EP2566515B1 - Innovative entdeckung von therapeutischen und diagnostischen mitteln sowie antikörperzusammensetzungen im zusammenhang mit proteinfragmenten von arginyl-trna-synthetasen - Google Patents
Innovative entdeckung von therapeutischen und diagnostischen mitteln sowie antikörperzusammensetzungen im zusammenhang mit proteinfragmenten von arginyl-trna-synthetasen Download PDFInfo
- Publication number
- EP2566515B1 EP2566515B1 EP11778118.7A EP11778118A EP2566515B1 EP 2566515 B1 EP2566515 B1 EP 2566515B1 EP 11778118 A EP11778118 A EP 11778118A EP 2566515 B1 EP2566515 B1 EP 2566515B1
- Authority
- EP
- European Patent Office
- Prior art keywords
- argrs
- polypeptide
- protein
- aars
- cell
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Active
Links
- 108090000623 proteins and genes Proteins 0.000 title claims description 349
- 102000004169 proteins and genes Human genes 0.000 title claims description 310
- 239000000203 mixture Substances 0.000 title claims description 91
- 230000001225 therapeutic effect Effects 0.000 title claims description 26
- 108010014885 Arginine-tRNA ligase Proteins 0.000 title description 419
- 239000012634 fragment Substances 0.000 title description 206
- 102100036131 Arginine-tRNA ligase, cytoplasmic Human genes 0.000 title 1
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 375
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 342
- 229920001184 polypeptide Polymers 0.000 claims description 321
- 210000004027 cell Anatomy 0.000 claims description 151
- 238000000034 method Methods 0.000 claims description 128
- 230000000694 effects Effects 0.000 claims description 120
- 239000002157 polynucleotide Substances 0.000 claims description 112
- 102000040430 polynucleotide Human genes 0.000 claims description 112
- 108091033319 polynucleotide Proteins 0.000 claims description 112
- 102000052866 Amino Acyl-tRNA Synthetases Human genes 0.000 claims description 97
- 108700028939 Amino Acyl-tRNA Synthetases Proteins 0.000 claims description 97
- 230000027455 binding Effects 0.000 claims description 96
- 241000282414 Homo sapiens Species 0.000 claims description 56
- 239000002773 nucleotide Substances 0.000 claims description 50
- 150000001875 compounds Chemical class 0.000 claims description 49
- 125000003729 nucleotide group Chemical group 0.000 claims description 48
- 230000001413 cellular effect Effects 0.000 claims description 44
- 239000002158 endotoxin Substances 0.000 claims description 40
- 239000011230 binding agent Substances 0.000 claims description 36
- 108091034117 Oligonucleotide Proteins 0.000 claims description 33
- 238000000338 in vitro Methods 0.000 claims description 20
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 claims description 18
- 238000012360 testing method Methods 0.000 claims description 18
- 230000000295 complement effect Effects 0.000 claims description 15
- 230000002797 proteolythic effect Effects 0.000 claims description 15
- 102000005962 receptors Human genes 0.000 claims description 15
- 238000001514 detection method Methods 0.000 claims description 13
- 108010001857 Cell Surface Receptors Proteins 0.000 claims description 12
- 230000003993 interaction Effects 0.000 claims description 12
- 230000017854 proteolysis Effects 0.000 claims description 10
- 239000000758 substrate Substances 0.000 claims description 9
- 230000008859 change Effects 0.000 claims description 8
- 239000008194 pharmaceutical composition Substances 0.000 claims description 8
- 230000008622 extracellular signaling Effects 0.000 claims description 6
- 239000012679 serum free medium Substances 0.000 claims description 4
- 208000012902 Nervous system disease Diseases 0.000 claims description 3
- 208000025966 Neurological disease Diseases 0.000 claims description 3
- 210000002901 mesenchymal stem cell Anatomy 0.000 claims description 3
- 239000003937 drug carrier Substances 0.000 claims description 2
- 239000001963 growth medium Substances 0.000 claims description 2
- 239000007787 solid Substances 0.000 claims description 2
- 102000006240 membrane receptors Human genes 0.000 claims 3
- 201000006474 Brain Ischemia Diseases 0.000 claims 1
- 208000036110 Neuroinflammatory disease Diseases 0.000 claims 1
- 230000004770 neurodegeneration Effects 0.000 claims 1
- 230000003959 neuroinflammation Effects 0.000 claims 1
- 238000002054 transplantation Methods 0.000 claims 1
- 102000002249 Arginine-tRNA Ligase Human genes 0.000 description 417
- 235000018102 proteins Nutrition 0.000 description 288
- 235000001014 amino acid Nutrition 0.000 description 122
- 229940024606 amino acid Drugs 0.000 description 115
- 150000001413 amino acids Chemical class 0.000 description 110
- 241000699666 Mus <mouse, genus> Species 0.000 description 101
- 108020004414 DNA Proteins 0.000 description 49
- 125000003275 alpha amino acid group Chemical group 0.000 description 44
- 230000014509 gene expression Effects 0.000 description 44
- 150000007523 nucleic acids Chemical class 0.000 description 44
- 239000003795 chemical substances by application Substances 0.000 description 40
- 239000013598 vector Substances 0.000 description 39
- 239000013615 primer Substances 0.000 description 38
- 230000004071 biological effect Effects 0.000 description 37
- 239000000523 sample Substances 0.000 description 36
- 125000000637 arginyl group Chemical group N[C@@H](CCCNC(N)=N)C(=O)* 0.000 description 35
- 239000004365 Protease Substances 0.000 description 33
- 102000035195 Peptidases Human genes 0.000 description 31
- 108091005804 Peptidases Proteins 0.000 description 31
- 102000039446 nucleic acids Human genes 0.000 description 28
- 108020004707 nucleic acids Proteins 0.000 description 28
- 238000006467 substitution reaction Methods 0.000 description 27
- 125000000539 amino acid group Chemical group 0.000 description 24
- 241000894007 species Species 0.000 description 24
- 238000003556 assay Methods 0.000 description 23
- 239000000427 antigen Substances 0.000 description 22
- 108091007433 antigens Proteins 0.000 description 22
- 102000036639 antigens Human genes 0.000 description 22
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 22
- 238000004519 manufacturing process Methods 0.000 description 21
- 239000012528 membrane Substances 0.000 description 21
- 239000013604 expression vector Substances 0.000 description 18
- 238000004949 mass spectrometry Methods 0.000 description 18
- 108020004999 messenger RNA Proteins 0.000 description 18
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 17
- 238000009396 hybridization Methods 0.000 description 17
- 230000014616 translation Effects 0.000 description 17
- -1 Arg amino acid Chemical class 0.000 description 16
- 201000010099 disease Diseases 0.000 description 16
- 238000000746 purification Methods 0.000 description 16
- 239000000126 substance Substances 0.000 description 16
- 238000005516 engineering process Methods 0.000 description 15
- 239000002245 particle Substances 0.000 description 15
- 150000003384 small molecules Chemical class 0.000 description 15
- 210000001519 tissue Anatomy 0.000 description 15
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 14
- 239000003153 chemical reaction reagent Substances 0.000 description 14
- 230000006870 function Effects 0.000 description 14
- 102000037865 fusion proteins Human genes 0.000 description 14
- 108020001507 fusion proteins Proteins 0.000 description 14
- 238000001727 in vivo Methods 0.000 description 14
- 229920000642 polymer Polymers 0.000 description 14
- 239000000047 product Substances 0.000 description 14
- 108020003175 receptors Proteins 0.000 description 14
- 102000004190 Enzymes Human genes 0.000 description 13
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 13
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 13
- 229940088598 enzyme Drugs 0.000 description 13
- 235000014304 histidine Nutrition 0.000 description 13
- 235000019419 proteases Nutrition 0.000 description 13
- 108090000790 Enzymes Proteins 0.000 description 12
- 108091028043 Nucleic acid sequence Proteins 0.000 description 12
- 238000004458 analytical method Methods 0.000 description 12
- 239000003814 drug Substances 0.000 description 12
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 12
- 230000035897 transcription Effects 0.000 description 12
- 238000013518 transcription Methods 0.000 description 12
- 231100000491 EC50 Toxicity 0.000 description 11
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 11
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 11
- 238000007792 addition Methods 0.000 description 11
- 239000002585 base Substances 0.000 description 11
- 210000004899 c-terminal region Anatomy 0.000 description 11
- 238000012217 deletion Methods 0.000 description 11
- 230000037430 deletion Effects 0.000 description 11
- 231100000673 dose–response relationship Toxicity 0.000 description 11
- 230000004927 fusion Effects 0.000 description 11
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 10
- 108060003951 Immunoglobulin Proteins 0.000 description 10
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 10
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 10
- 230000015572 biosynthetic process Effects 0.000 description 10
- 229940079593 drug Drugs 0.000 description 10
- 238000007876 drug discovery Methods 0.000 description 10
- 102000018358 immunoglobulin Human genes 0.000 description 10
- 230000004048 modification Effects 0.000 description 10
- 238000012986 modification Methods 0.000 description 10
- 238000012545 processing Methods 0.000 description 10
- 238000011160 research Methods 0.000 description 10
- 230000001177 retroviral effect Effects 0.000 description 10
- 241000894006 Bacteria Species 0.000 description 9
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 9
- 102000000844 Cell Surface Receptors Human genes 0.000 description 9
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 9
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 9
- 241000700605 Viruses Species 0.000 description 9
- 230000002776 aggregation Effects 0.000 description 9
- 238000004220 aggregation Methods 0.000 description 9
- 229940098773 bovine serum albumin Drugs 0.000 description 9
- 239000000975 dye Substances 0.000 description 9
- 230000001404 mediated effect Effects 0.000 description 9
- 229920001223 polyethylene glycol Polymers 0.000 description 9
- 239000002987 primer (paints) Substances 0.000 description 9
- 230000008569 process Effects 0.000 description 9
- 210000002966 serum Anatomy 0.000 description 9
- 239000002904 solvent Substances 0.000 description 9
- 238000013519 translation Methods 0.000 description 9
- 108091026890 Coding region Proteins 0.000 description 8
- 108700022150 Designed Ankyrin Repeat Proteins Proteins 0.000 description 8
- 241000282412 Homo Species 0.000 description 8
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 8
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 8
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 8
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 8
- 241001465754 Metazoa Species 0.000 description 8
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 8
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 8
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 8
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 8
- 239000004473 Threonine Substances 0.000 description 8
- 230000001580 bacterial effect Effects 0.000 description 8
- 238000004166 bioassay Methods 0.000 description 8
- 239000003446 ligand Substances 0.000 description 8
- 235000013930 proline Nutrition 0.000 description 8
- 150000003839 salts Chemical class 0.000 description 8
- 235000004400 serine Nutrition 0.000 description 8
- 238000003786 synthesis reaction Methods 0.000 description 8
- 235000008521 threonine Nutrition 0.000 description 8
- 238000011282 treatment Methods 0.000 description 8
- 238000005406 washing Methods 0.000 description 8
- 239000004475 Arginine Substances 0.000 description 7
- 108020004635 Complementary DNA Proteins 0.000 description 7
- 238000002965 ELISA Methods 0.000 description 7
- 241000588724 Escherichia coli Species 0.000 description 7
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 7
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 7
- 102000003960 Ligases Human genes 0.000 description 7
- 108090000364 Ligases Proteins 0.000 description 7
- 239000004472 Lysine Substances 0.000 description 7
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 7
- 108020004566 Transfer RNA Proteins 0.000 description 7
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 7
- 230000000692 anti-sense effect Effects 0.000 description 7
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 7
- 235000009697 arginine Nutrition 0.000 description 7
- 238000010804 cDNA synthesis Methods 0.000 description 7
- 238000012512 characterization method Methods 0.000 description 7
- 238000006243 chemical reaction Methods 0.000 description 7
- 238000010367 cloning Methods 0.000 description 7
- 239000002299 complementary DNA Substances 0.000 description 7
- 239000000306 component Substances 0.000 description 7
- 239000013078 crystal Substances 0.000 description 7
- 238000012350 deep sequencing Methods 0.000 description 7
- 235000013922 glutamic acid Nutrition 0.000 description 7
- 229960002989 glutamic acid Drugs 0.000 description 7
- 239000004220 glutamic acid Substances 0.000 description 7
- 238000004128 high performance liquid chromatography Methods 0.000 description 7
- 238000003780 insertion Methods 0.000 description 7
- 230000037431 insertion Effects 0.000 description 7
- 235000018977 lysine Nutrition 0.000 description 7
- 239000011159 matrix material Substances 0.000 description 7
- 239000013612 plasmid Substances 0.000 description 7
- 239000002096 quantum dot Substances 0.000 description 7
- 230000004044 response Effects 0.000 description 7
- 238000012552 review Methods 0.000 description 7
- 230000028327 secretion Effects 0.000 description 7
- 108020004705 Codon Proteins 0.000 description 6
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 6
- 239000004471 Glycine Substances 0.000 description 6
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 6
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 6
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 6
- 108010079855 Peptide Aptamers Proteins 0.000 description 6
- 102000001708 Protein Isoforms Human genes 0.000 description 6
- 108010029485 Protein Isoforms Proteins 0.000 description 6
- 210000001744 T-lymphocyte Anatomy 0.000 description 6
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 6
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical group [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 6
- QTBSBXVTEAMEQO-UHFFFAOYSA-N acetic acid Substances CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 6
- 230000003321 amplification Effects 0.000 description 6
- 238000013459 approach Methods 0.000 description 6
- 229910052799 carbon Inorganic materials 0.000 description 6
- 238000000423 cell based assay Methods 0.000 description 6
- 230000024245 cell differentiation Effects 0.000 description 6
- 208000035475 disorder Diseases 0.000 description 6
- 230000005847 immunogenicity Effects 0.000 description 6
- 210000004962 mammalian cell Anatomy 0.000 description 6
- 239000002609 medium Substances 0.000 description 6
- 238000003199 nucleic acid amplification method Methods 0.000 description 6
- 239000000816 peptidomimetic Substances 0.000 description 6
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 6
- 229910052725 zinc Inorganic materials 0.000 description 6
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 5
- 241000196324 Embryophyta Species 0.000 description 5
- 102000005720 Glutathione transferase Human genes 0.000 description 5
- 108010070675 Glutathione transferase Proteins 0.000 description 5
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 5
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 5
- 206010061218 Inflammation Diseases 0.000 description 5
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 5
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 5
- 125000000174 L-prolyl group Chemical group [H]N1C([H])([H])C([H])([H])C([H])([H])[C@@]1([H])C(*)=O 0.000 description 5
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 5
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 5
- 108091027974 Mature messenger RNA Proteins 0.000 description 5
- 108020004511 Recombinant DNA Proteins 0.000 description 5
- 241000283984 Rodentia Species 0.000 description 5
- 108091081024 Start codon Proteins 0.000 description 5
- COYHRQWNJDJCNA-NUJDXYNKSA-N Thr-Thr-Thr Chemical compound C[C@@H](O)[C@H](N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H]([C@@H](C)O)C(O)=O COYHRQWNJDJCNA-NUJDXYNKSA-N 0.000 description 5
- 239000000556 agonist Substances 0.000 description 5
- 235000004279 alanine Nutrition 0.000 description 5
- 239000005557 antagonist Substances 0.000 description 5
- 125000003118 aryl group Chemical group 0.000 description 5
- 235000009582 asparagine Nutrition 0.000 description 5
- 229960001230 asparagine Drugs 0.000 description 5
- 235000003704 aspartic acid Nutrition 0.000 description 5
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 5
- 238000004422 calculation algorithm Methods 0.000 description 5
- 230000030833 cell death Effects 0.000 description 5
- 238000002659 cell therapy Methods 0.000 description 5
- 235000018417 cysteine Nutrition 0.000 description 5
- 239000007850 fluorescent dye Substances 0.000 description 5
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 5
- 235000004554 glutamine Nutrition 0.000 description 5
- 230000001965 increasing effect Effects 0.000 description 5
- 230000001939 inductive effect Effects 0.000 description 5
- 230000004054 inflammatory process Effects 0.000 description 5
- 229960000310 isoleucine Drugs 0.000 description 5
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 5
- 229920006008 lipopolysaccharide Polymers 0.000 description 5
- 239000003550 marker Substances 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- 238000002703 mutagenesis Methods 0.000 description 5
- 231100000350 mutagenesis Toxicity 0.000 description 5
- 230000035772 mutation Effects 0.000 description 5
- 238000003752 polymerase chain reaction Methods 0.000 description 5
- 230000001105 regulatory effect Effects 0.000 description 5
- 238000012216 screening Methods 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 230000008685 targeting Effects 0.000 description 5
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 5
- 235000002374 tyrosine Nutrition 0.000 description 5
- 239000004474 valine Substances 0.000 description 5
- 230000003612 virological effect Effects 0.000 description 5
- 239000011701 zinc Substances 0.000 description 5
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- 108091023037 Aptamer Proteins 0.000 description 4
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 4
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 4
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 4
- 241000124008 Mammalia Species 0.000 description 4
- 102000005741 Metalloproteases Human genes 0.000 description 4
- 108010006035 Metalloproteases Proteins 0.000 description 4
- 102000016387 Pancreatic elastase Human genes 0.000 description 4
- 108010067372 Pancreatic elastase Proteins 0.000 description 4
- 108010043958 Peptoids Proteins 0.000 description 4
- 102100026126 Proline-tRNA ligase Human genes 0.000 description 4
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 4
- 241000714474 Rous sarcoma virus Species 0.000 description 4
- 238000012300 Sequence Analysis Methods 0.000 description 4
- 102000012479 Serine Proteases Human genes 0.000 description 4
- 108010022999 Serine Proteases Proteins 0.000 description 4
- BQCADISMDOOEFD-UHFFFAOYSA-N Silver Chemical compound [Ag] BQCADISMDOOEFD-UHFFFAOYSA-N 0.000 description 4
- 230000002378 acidificating effect Effects 0.000 description 4
- 230000004075 alteration Effects 0.000 description 4
- 150000001408 amides Chemical class 0.000 description 4
- 230000006229 amino acid addition Effects 0.000 description 4
- 238000012801 analytical assay Methods 0.000 description 4
- 230000033115 angiogenesis Effects 0.000 description 4
- 230000006907 apoptotic process Effects 0.000 description 4
- 239000012736 aqueous medium Substances 0.000 description 4
- 239000007864 aqueous solution Substances 0.000 description 4
- 150000001720 carbohydrates Chemical class 0.000 description 4
- 235000014633 carbohydrates Nutrition 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 230000004663 cell proliferation Effects 0.000 description 4
- 230000005754 cellular signaling Effects 0.000 description 4
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 4
- 230000016396 cytokine production Effects 0.000 description 4
- 108010057085 cytokine receptors Proteins 0.000 description 4
- 102000003675 cytokine receptors Human genes 0.000 description 4
- 230000001086 cytosolic effect Effects 0.000 description 4
- 230000034994 death Effects 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 239000003623 enhancer Substances 0.000 description 4
- 230000002255 enzymatic effect Effects 0.000 description 4
- 210000003527 eukaryotic cell Anatomy 0.000 description 4
- 230000009368 gene silencing by RNA Effects 0.000 description 4
- 238000001415 gene therapy Methods 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 4
- 230000002209 hydrophobic effect Effects 0.000 description 4
- 210000003734 kidney Anatomy 0.000 description 4
- 210000003292 kidney cell Anatomy 0.000 description 4
- 238000002372 labelling Methods 0.000 description 4
- 150000002632 lipids Chemical class 0.000 description 4
- 229930182817 methionine Natural products 0.000 description 4
- 238000002493 microarray Methods 0.000 description 4
- 238000010369 molecular cloning Methods 0.000 description 4
- 239000000178 monomer Substances 0.000 description 4
- 239000002105 nanoparticle Substances 0.000 description 4
- 210000000056 organ Anatomy 0.000 description 4
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 4
- 239000002243 precursor Substances 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 235000019833 protease Nutrition 0.000 description 4
- 229910052709 silver Inorganic materials 0.000 description 4
- 239000004332 silver Substances 0.000 description 4
- 238000005556 structure-activity relationship Methods 0.000 description 4
- 208000024891 symptom Diseases 0.000 description 4
- 230000009897 systematic effect Effects 0.000 description 4
- 210000002845 virion Anatomy 0.000 description 4
- 102000035101 Aspartic proteases Human genes 0.000 description 3
- 108091005502 Aspartic proteases Proteins 0.000 description 3
- 108010032088 Calpain Proteins 0.000 description 3
- 102000007590 Calpain Human genes 0.000 description 3
- 102000014914 Carrier Proteins Human genes 0.000 description 3
- 108090000746 Chymosin Proteins 0.000 description 3
- 108090000317 Chymotrypsin Proteins 0.000 description 3
- 102000005927 Cysteine Proteases Human genes 0.000 description 3
- 108010005843 Cysteine Proteases Proteins 0.000 description 3
- 102000053602 DNA Human genes 0.000 description 3
- 238000012286 ELISA Assay Methods 0.000 description 3
- 241000238631 Hexapoda Species 0.000 description 3
- 101001027128 Homo sapiens Fibronectin Proteins 0.000 description 3
- 241000725303 Human immunodeficiency virus Species 0.000 description 3
- 108010004098 Leucyl aminopeptidase Proteins 0.000 description 3
- 102000002704 Leucyl aminopeptidase Human genes 0.000 description 3
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 3
- 108090000526 Papain Proteins 0.000 description 3
- 108090000284 Pepsin A Proteins 0.000 description 3
- 102000057297 Pepsin A Human genes 0.000 description 3
- 101710182846 Polyhedrin Proteins 0.000 description 3
- 108010076504 Protein Sorting Signals Proteins 0.000 description 3
- 108091028664 Ribonucleotide Proteins 0.000 description 3
- 108090001109 Thermolysin Proteins 0.000 description 3
- 241000723873 Tobacco mosaic virus Species 0.000 description 3
- 102000004142 Trypsin Human genes 0.000 description 3
- 108090000631 Trypsin Proteins 0.000 description 3
- 230000001594 aberrant effect Effects 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- DZBUGLKDJFMEHC-UHFFFAOYSA-N acridine Chemical compound C1=CC=CC2=CC3=CC=CC=C3N=C21 DZBUGLKDJFMEHC-UHFFFAOYSA-N 0.000 description 3
- 230000002411 adverse Effects 0.000 description 3
- 150000001412 amines Chemical class 0.000 description 3
- 238000013103 analytical ultracentrifugation Methods 0.000 description 3
- 210000004102 animal cell Anatomy 0.000 description 3
- 239000000074 antisense oligonucleotide Substances 0.000 description 3
- 238000012230 antisense oligonucleotides Methods 0.000 description 3
- 108091008324 binding proteins Proteins 0.000 description 3
- 229960000074 biopharmaceutical Drugs 0.000 description 3
- 210000004556 brain Anatomy 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 230000012292 cell migration Effects 0.000 description 3
- 230000004700 cellular uptake Effects 0.000 description 3
- 229960002376 chymotrypsin Drugs 0.000 description 3
- 238000010168 coupling process Methods 0.000 description 3
- 238000005859 coupling reaction Methods 0.000 description 3
- 125000004122 cyclic group Chemical group 0.000 description 3
- 230000003013 cytotoxicity Effects 0.000 description 3
- 231100000135 cytotoxicity Toxicity 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 239000000539 dimer Substances 0.000 description 3
- 238000007824 enzymatic assay Methods 0.000 description 3
- 239000000284 extract Substances 0.000 description 3
- 230000002349 favourable effect Effects 0.000 description 3
- 230000013595 glycosylation Effects 0.000 description 3
- 238000006206 glycosylation reaction Methods 0.000 description 3
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 3
- 229910052737 gold Inorganic materials 0.000 description 3
- 239000010931 gold Substances 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 230000001900 immune effect Effects 0.000 description 3
- 230000001976 improved effect Effects 0.000 description 3
- 230000006872 improvement Effects 0.000 description 3
- 238000010348 incorporation Methods 0.000 description 3
- 208000015181 infectious disease Diseases 0.000 description 3
- 208000027866 inflammatory disease Diseases 0.000 description 3
- 230000009878 intermolecular interaction Effects 0.000 description 3
- 238000004255 ion exchange chromatography Methods 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 230000000670 limiting effect Effects 0.000 description 3
- 125000005647 linker group Chemical group 0.000 description 3
- 239000002502 liposome Substances 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 description 3
- 230000007935 neutral effect Effects 0.000 description 3
- 108091008104 nucleic acid aptamers Proteins 0.000 description 3
- 238000004806 packaging method and process Methods 0.000 description 3
- 229940055729 papain Drugs 0.000 description 3
- 235000019834 papain Nutrition 0.000 description 3
- 230000006320 pegylation Effects 0.000 description 3
- 229940111202 pepsin Drugs 0.000 description 3
- 238000010647 peptide synthesis reaction Methods 0.000 description 3
- 230000004481 post-translational protein modification Effects 0.000 description 3
- 230000001323 posttranslational effect Effects 0.000 description 3
- 238000001243 protein synthesis Methods 0.000 description 3
- 238000002271 resection Methods 0.000 description 3
- 230000002441 reversible effect Effects 0.000 description 3
- 239000002336 ribonucleotide Substances 0.000 description 3
- 125000002652 ribonucleotide group Chemical group 0.000 description 3
- 238000012163 sequencing technique Methods 0.000 description 3
- 238000001542 size-exclusion chromatography Methods 0.000 description 3
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 3
- ABZLKHKQJHEPAX-UHFFFAOYSA-N tetramethylrhodamine Chemical compound C=12C=CC(N(C)C)=CC2=[O+]C2=CC(N(C)C)=CC=C2C=1C1=CC=CC=C1C([O-])=O ABZLKHKQJHEPAX-UHFFFAOYSA-N 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 230000002103 transcriptional effect Effects 0.000 description 3
- 230000001052 transient effect Effects 0.000 description 3
- 239000012588 trypsin Substances 0.000 description 3
- 241000701161 unidentified adenovirus Species 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- WQZIDRAQTRIQDX-UHFFFAOYSA-N 6-carboxy-x-rhodamine Chemical compound OC(=O)C1=CC=C(C([O-])=O)C=C1C(C1=CC=2CCCN3CCCC(C=23)=C1O1)=C2C1=C(CCC1)C3=[N+]1CCCC3=C2 WQZIDRAQTRIQDX-UHFFFAOYSA-N 0.000 description 2
- BZTDTCNHAFUJOG-UHFFFAOYSA-N 6-carboxyfluorescein Chemical compound C12=CC=C(O)C=C2OC2=CC(O)=CC=C2C11OC(=O)C2=CC=C(C(=O)O)C=C21 BZTDTCNHAFUJOG-UHFFFAOYSA-N 0.000 description 2
- 102000006268 Alanine-tRNA ligase Human genes 0.000 description 2
- 108010058060 Alanine-tRNA ligase Proteins 0.000 description 2
- IKYJCHYORFJFRR-UHFFFAOYSA-N Alexa Fluor 350 Chemical compound O=C1OC=2C=C(N)C(S(O)(=O)=O)=CC=2C(C)=C1CC(=O)ON1C(=O)CCC1=O IKYJCHYORFJFRR-UHFFFAOYSA-N 0.000 description 2
- WHVNXSBKJGAXKU-UHFFFAOYSA-N Alexa Fluor 532 Chemical compound [H+].[H+].CC1(C)C(C)NC(C(=C2OC3=C(C=4C(C(C(C)N=4)(C)C)=CC3=3)S([O-])(=O)=O)S([O-])(=O)=O)=C1C=C2C=3C(C=C1)=CC=C1C(=O)ON1C(=O)CCC1=O WHVNXSBKJGAXKU-UHFFFAOYSA-N 0.000 description 2
- ZAINTDRBUHCDPZ-UHFFFAOYSA-M Alexa Fluor 546 Chemical compound [H+].[Na+].CC1CC(C)(C)NC(C(=C2OC3=C(C4=NC(C)(C)CC(C)C4=CC3=3)S([O-])(=O)=O)S([O-])(=O)=O)=C1C=C2C=3C(C(=C(Cl)C=1Cl)C(O)=O)=C(Cl)C=1SCC(=O)NCCCCCC(=O)ON1C(=O)CCC1=O ZAINTDRBUHCDPZ-UHFFFAOYSA-M 0.000 description 2
- 101000640990 Arabidopsis thaliana Tryptophan-tRNA ligase, chloroplastic/mitochondrial Proteins 0.000 description 2
- 101000787278 Arabidopsis thaliana Valine-tRNA ligase, chloroplastic/mitochondrial 2 Proteins 0.000 description 2
- 101000787296 Arabidopsis thaliana Valine-tRNA ligase, mitochondrial 1 Proteins 0.000 description 2
- 102000003924 Asparagine-tRNA ligases Human genes 0.000 description 2
- 108090000314 Asparagine-tRNA ligases Proteins 0.000 description 2
- 102000012951 Aspartate-tRNA Ligase Human genes 0.000 description 2
- 108010065272 Aspartate-tRNA ligase Proteins 0.000 description 2
- 108090001008 Avidin Proteins 0.000 description 2
- 206010006187 Breast cancer Diseases 0.000 description 2
- 108010004032 Bromelains Proteins 0.000 description 2
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 2
- 208000024172 Cardiovascular disease Diseases 0.000 description 2
- 108090000426 Caspase-1 Proteins 0.000 description 2
- 102000005600 Cathepsins Human genes 0.000 description 2
- 108010084457 Cathepsins Proteins 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 2
- 108091035707 Consensus sequence Proteins 0.000 description 2
- 102000004403 Cysteine-tRNA ligases Human genes 0.000 description 2
- 108090000918 Cysteine-tRNA ligases Proteins 0.000 description 2
- 101000787280 Dictyostelium discoideum Probable valine-tRNA ligase, mitochondrial Proteins 0.000 description 2
- SHIBSTMRCDJXLN-UHFFFAOYSA-N Digoxigenin Natural products C1CC(C2C(C3(C)CCC(O)CC3CC2)CC2O)(O)C2(C)C1C1=CC(=O)OC1 SHIBSTMRCDJXLN-UHFFFAOYSA-N 0.000 description 2
- 241001635598 Enicostema Species 0.000 description 2
- 108010074860 Factor Xa Proteins 0.000 description 2
- 108010015514 Glutamate-tRNA ligase Proteins 0.000 description 2
- 102100024977 Glutamine-tRNA ligase Human genes 0.000 description 2
- 108010024636 Glutathione Proteins 0.000 description 2
- 108010051724 Glycine-tRNA Ligase Proteins 0.000 description 2
- 102000019220 Glycyl-tRNA synthetases Human genes 0.000 description 2
- 102000001398 Granzyme Human genes 0.000 description 2
- 108060005986 Granzyme Proteins 0.000 description 2
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 2
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 2
- NYHBQMYGNKIUIF-UUOKFMHZSA-N Guanosine Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O NYHBQMYGNKIUIF-UUOKFMHZSA-N 0.000 description 2
- 102000006354 HLA-DR Antigens Human genes 0.000 description 2
- 108010058597 HLA-DR Antigens Proteins 0.000 description 2
- 241000713858 Harvey murine sarcoma virus Species 0.000 description 2
- 108010093488 His-His-His-His-His-His Proteins 0.000 description 2
- 102000029746 Histidine-tRNA Ligase Human genes 0.000 description 2
- 101710177011 Histidine-tRNA ligase, cytoplasmic Proteins 0.000 description 2
- 101100479034 Homo sapiens AARS1 gene Proteins 0.000 description 2
- 101000874860 Homo sapiens Arginine-tRNA ligase, cytoplasmic Proteins 0.000 description 2
- 101000874919 Homo sapiens Probable arginine-tRNA ligase, mitochondrial Proteins 0.000 description 2
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 2
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 2
- SIKJAQJRHWYJAI-UHFFFAOYSA-N Indole Chemical compound C1=CC=C2NC=CC2=C1 SIKJAQJRHWYJAI-UHFFFAOYSA-N 0.000 description 2
- 108091092195 Intron Proteins 0.000 description 2
- 102000029793 Isoleucine-tRNA ligase Human genes 0.000 description 2
- 101710176147 Isoleucine-tRNA ligase, cytoplasmic Proteins 0.000 description 2
- 102000001399 Kallikrein Human genes 0.000 description 2
- 108060005987 Kallikrein Proteins 0.000 description 2
- 108010071170 Leucine-tRNA ligase Proteins 0.000 description 2
- 102100023342 Leucine-tRNA ligase, mitochondrial Human genes 0.000 description 2
- 241000239218 Limulus Species 0.000 description 2
- 239000000232 Lipid Bilayer Substances 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 102000017737 Lysine-tRNA Ligase Human genes 0.000 description 2
- 108010092041 Lysine-tRNA Ligase Proteins 0.000 description 2
- 102000000362 Methionyl-tRNA synthetases Human genes 0.000 description 2
- 108050008914 Methionyl-tRNA synthetases Proteins 0.000 description 2
- 241000713869 Moloney murine leukemia virus Species 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- 108010049175 N-substituted Glycines Proteins 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 102000002798 Phenylalanine-tRNA Ligase Human genes 0.000 description 2
- 108010004478 Phenylalanine-tRNA Ligase Proteins 0.000 description 2
- 239000004372 Polyvinyl alcohol Substances 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 101710096715 Probable histidine-tRNA ligase, cytoplasmic Proteins 0.000 description 2
- 101710149031 Probable isoleucine-tRNA ligase, cytoplasmic Proteins 0.000 description 2
- 101710146427 Probable tyrosine-tRNA ligase, cytoplasmic Proteins 0.000 description 2
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 2
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 2
- 108010030161 Serine-tRNA ligase Proteins 0.000 description 2
- 102100040516 Serine-tRNA ligase, cytoplasmic Human genes 0.000 description 2
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 241000256251 Spodoptera frugiperda Species 0.000 description 2
- 108090000787 Subtilisin Proteins 0.000 description 2
- 102000001618 Threonine-tRNA Ligase Human genes 0.000 description 2
- 108010029287 Threonine-tRNA ligase Proteins 0.000 description 2
- 108090000190 Thrombin Proteins 0.000 description 2
- 241000255993 Trichoplusia ni Species 0.000 description 2
- 102000002501 Tryptophan-tRNA Ligase Human genes 0.000 description 2
- 102000018378 Tyrosine-tRNA ligase Human genes 0.000 description 2
- 101710107268 Tyrosine-tRNA ligase, mitochondrial Proteins 0.000 description 2
- DRTQHJPVMGBUCF-XVFCMESISA-N Uridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-XVFCMESISA-N 0.000 description 2
- 102000013625 Valine-tRNA Ligase Human genes 0.000 description 2
- 230000021736 acetylation Effects 0.000 description 2
- 238000006640 acetylation reaction Methods 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 2
- 238000012382 advanced drug delivery Methods 0.000 description 2
- 238000001261 affinity purification Methods 0.000 description 2
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 230000003042 antagnostic effect Effects 0.000 description 2
- 210000000612 antigen-presenting cell Anatomy 0.000 description 2
- RASZIXQTZOARSV-BDPUVYQTSA-N astacin Chemical compound CC=1C(=O)C(=O)CC(C)(C)C=1/C=C/C(/C)=C/C=C/C(/C)=C/C=C/C=C(C)C=CC=C(C)C=CC1=C(C)C(=O)C(=O)CC1(C)C RASZIXQTZOARSV-BDPUVYQTSA-N 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000003115 biocidal effect Effects 0.000 description 2
- 238000003766 bioinformatics method Methods 0.000 description 2
- 239000013060 biological fluid Substances 0.000 description 2
- 230000008827 biological function Effects 0.000 description 2
- 230000008236 biological pathway Effects 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 230000008499 blood brain barrier function Effects 0.000 description 2
- 210000001218 blood-brain barrier Anatomy 0.000 description 2
- 235000019835 bromelain Nutrition 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 238000006555 catalytic reaction Methods 0.000 description 2
- 230000019522 cellular metabolic process Effects 0.000 description 2
- 238000007385 chemical modification Methods 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- 230000015271 coagulation Effects 0.000 description 2
- 238000005345 coagulation Methods 0.000 description 2
- 108091036078 conserved sequence Proteins 0.000 description 2
- 239000013068 control sample Substances 0.000 description 2
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 2
- SUYVUBYJARFZHO-UHFFFAOYSA-N dATP Natural products C1=NC=2C(N)=NC=NC=2N1C1CC(O)C(COP(O)(=O)OP(O)(=O)OP(O)(O)=O)O1 SUYVUBYJARFZHO-UHFFFAOYSA-N 0.000 description 2
- SUYVUBYJARFZHO-RRKCRQDMSA-N dATP Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@H]1C[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OP(O)(O)=O)O1 SUYVUBYJARFZHO-RRKCRQDMSA-N 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- 239000000032 diagnostic agent Substances 0.000 description 2
- 229940039227 diagnostic agent Drugs 0.000 description 2
- QONQRTHLHBTMGP-UHFFFAOYSA-N digitoxigenin Natural products CC12CCC(C3(CCC(O)CC3CC3)C)C3C11OC1CC2C1=CC(=O)OC1 QONQRTHLHBTMGP-UHFFFAOYSA-N 0.000 description 2
- SHIBSTMRCDJXLN-KCZCNTNESA-N digoxigenin Chemical compound C1([C@@H]2[C@@]3([C@@](CC2)(O)[C@H]2[C@@H]([C@@]4(C)CC[C@H](O)C[C@H]4CC2)C[C@H]3O)C)=CC(=O)OC1 SHIBSTMRCDJXLN-KCZCNTNESA-N 0.000 description 2
- 238000009509 drug development Methods 0.000 description 2
- 239000003596 drug target Substances 0.000 description 2
- 238000002296 dynamic light scattering Methods 0.000 description 2
- 238000005538 encapsulation Methods 0.000 description 2
- 235000020776 essential amino acid Nutrition 0.000 description 2
- 239000003797 essential amino acid Substances 0.000 description 2
- 238000001914 filtration Methods 0.000 description 2
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 2
- 238000002866 fluorescence resonance energy transfer Methods 0.000 description 2
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 2
- 238000004108 freeze drying Methods 0.000 description 2
- 108010051239 glutaminyl-tRNA synthetase Proteins 0.000 description 2
- 229960003180 glutathione Drugs 0.000 description 2
- 239000005090 green fluorescent protein Substances 0.000 description 2
- 210000002443 helper t lymphocyte Anatomy 0.000 description 2
- 208000014951 hematologic disease Diseases 0.000 description 2
- 230000011132 hemopoiesis Effects 0.000 description 2
- 239000000833 heterodimer Substances 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 238000001597 immobilized metal affinity chromatography Methods 0.000 description 2
- 238000003119 immunoblot Methods 0.000 description 2
- 230000002163 immunogen Effects 0.000 description 2
- 238000000126 in silico method Methods 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 150000002500 ions Chemical class 0.000 description 2
- 231100000636 lethal dose Toxicity 0.000 description 2
- 238000001638 lipofection Methods 0.000 description 2
- 210000005229 liver cell Anatomy 0.000 description 2
- 239000006166 lysate Substances 0.000 description 2
- 230000002132 lysosomal effect Effects 0.000 description 2
- 229920002521 macromolecule Polymers 0.000 description 2
- 208000030159 metabolic disease Diseases 0.000 description 2
- 229910052751 metal Inorganic materials 0.000 description 2
- 239000002184 metal Substances 0.000 description 2
- 244000005700 microbiome Species 0.000 description 2
- 230000002438 mitochondrial effect Effects 0.000 description 2
- 239000003607 modifier Substances 0.000 description 2
- 210000000440 neutrophil Anatomy 0.000 description 2
- 229910052759 nickel Inorganic materials 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 231100000706 no observed effect level Toxicity 0.000 description 2
- 238000007899 nucleic acid hybridization Methods 0.000 description 2
- 239000002777 nucleoside Substances 0.000 description 2
- 239000002751 oligonucleotide probe Substances 0.000 description 2
- 238000002515 oligonucleotide synthesis Methods 0.000 description 2
- 230000003287 optical effect Effects 0.000 description 2
- 238000005457 optimization Methods 0.000 description 2
- 150000002894 organic compounds Chemical class 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 230000001717 pathogenic effect Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 239000012071 phase Substances 0.000 description 2
- 125000002467 phosphate group Chemical group [H]OP(=O)(O[H])O[*] 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 230000001766 physiological effect Effects 0.000 description 2
- 230000035790 physiological processes and functions Effects 0.000 description 2
- 230000008488 polyadenylation Effects 0.000 description 2
- 229920002451 polyvinyl alcohol Polymers 0.000 description 2
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 2
- 108010042589 prolyl T RNA synthetase Proteins 0.000 description 2
- 238000001742 protein purification Methods 0.000 description 2
- 230000006337 proteolytic cleavage Effects 0.000 description 2
- BBEAQIROQSPTKN-UHFFFAOYSA-N pyrene Chemical compound C1=CC=C2C=CC3=CC=CC4=CC=C1C2=C43 BBEAQIROQSPTKN-UHFFFAOYSA-N 0.000 description 2
- 238000003127 radioimmunoassay Methods 0.000 description 2
- 238000010188 recombinant method Methods 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 108091008146 restriction endonucleases Proteins 0.000 description 2
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 2
- 230000007017 scission Effects 0.000 description 2
- 239000004017 serum-free culture medium Substances 0.000 description 2
- 238000003998 size exclusion chromatography high performance liquid chromatography Methods 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 125000001424 substituent group Chemical group 0.000 description 2
- 238000004114 suspension culture Methods 0.000 description 2
- MPLHNVLQVRSVEE-UHFFFAOYSA-N texas red Chemical compound [O-]S(=O)(=O)C1=CC(S(Cl)(=O)=O)=CC=C1C(C1=CC=2CCCN3CCCC(C=23)=C1O1)=C2C1=C(CCC1)C3=[N+]1CCCC3=C2 MPLHNVLQVRSVEE-UHFFFAOYSA-N 0.000 description 2
- 229960004072 thrombin Drugs 0.000 description 2
- 239000003053 toxin Substances 0.000 description 2
- 231100000765 toxin Toxicity 0.000 description 2
- 108700012359 toxins Proteins 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- 239000001226 triphosphate Substances 0.000 description 2
- 235000011178 triphosphate Nutrition 0.000 description 2
- 241000701447 unidentified baculovirus Species 0.000 description 2
- 238000011144 upstream manufacturing Methods 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- 229920003169 water-soluble polymer Polymers 0.000 description 2
- VEVRNHHLCPGNDU-MUGJNUQGSA-N (2s)-2-amino-5-[1-[(5s)-5-amino-5-carboxypentyl]-3,5-bis[(3s)-3-amino-3-carboxypropyl]pyridin-1-ium-4-yl]pentanoate Chemical compound OC(=O)[C@@H](N)CCCC[N+]1=CC(CC[C@H](N)C(O)=O)=C(CCC[C@H](N)C([O-])=O)C(CC[C@H](N)C(O)=O)=C1 VEVRNHHLCPGNDU-MUGJNUQGSA-N 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 1
- WNXJIVFYUVYPPR-UHFFFAOYSA-N 1,3-dioxolane Chemical compound C1COCO1 WNXJIVFYUVYPPR-UHFFFAOYSA-N 0.000 description 1
- UUUHXMGGBIUAPW-UHFFFAOYSA-N 1-[1-[2-[[5-amino-2-[[1-[5-(diaminomethylideneamino)-2-[[1-[3-(1h-indol-3-yl)-2-[(5-oxopyrrolidine-2-carbonyl)amino]propanoyl]pyrrolidine-2-carbonyl]amino]pentanoyl]pyrrolidine-2-carbonyl]amino]-5-oxopentanoyl]amino]-3-methylpentanoyl]pyrrolidine-2-carbon Chemical compound C1CCC(C(=O)N2C(CCC2)C(O)=O)N1C(=O)C(C(C)CC)NC(=O)C(CCC(N)=O)NC(=O)C1CCCN1C(=O)C(CCCN=C(N)N)NC(=O)C1CCCN1C(=O)C(CC=1C2=CC=CC=C2NC=1)NC(=O)C1CCC(=O)N1 UUUHXMGGBIUAPW-UHFFFAOYSA-N 0.000 description 1
- RUFPHBVGCFYCNW-UHFFFAOYSA-N 1-naphthylamine Chemical compound C1=CC=C2C(N)=CC=CC2=C1 RUFPHBVGCFYCNW-UHFFFAOYSA-N 0.000 description 1
- OWEGMIWEEQEYGQ-UHFFFAOYSA-N 100676-05-9 Natural products OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(OC2C(OC(O)C(O)C2O)CO)O1 OWEGMIWEEQEYGQ-UHFFFAOYSA-N 0.000 description 1
- SVUOLADPCWQTTE-UHFFFAOYSA-N 1h-1,2-benzodiazepine Chemical compound N1N=CC=CC2=CC=CC=C12 SVUOLADPCWQTTE-UHFFFAOYSA-N 0.000 description 1
- 150000003923 2,5-pyrrolediones Chemical class 0.000 description 1
- CUVGUPIVTLGRGI-UHFFFAOYSA-N 4-(3-phosphonopropyl)piperazine-2-carboxylic acid Chemical compound OC(=O)C1CN(CCCP(O)(O)=O)CCN1 CUVGUPIVTLGRGI-UHFFFAOYSA-N 0.000 description 1
- LKDMKWNDBAVNQZ-UHFFFAOYSA-N 4-[[1-[[1-[2-[[1-(4-nitroanilino)-1-oxo-3-phenylpropan-2-yl]carbamoyl]pyrrolidin-1-yl]-1-oxopropan-2-yl]amino]-1-oxopropan-2-yl]amino]-4-oxobutanoic acid Chemical compound OC(=O)CCC(=O)NC(C)C(=O)NC(C)C(=O)N1CCCC1C(=O)NC(C(=O)NC=1C=CC(=CC=1)[N+]([O-])=O)CC1=CC=CC=C1 LKDMKWNDBAVNQZ-UHFFFAOYSA-N 0.000 description 1
- XWHHYOYVRVGJJY-UHFFFAOYSA-N 4-fluorophenylalanine Chemical compound OC(=O)C(N)CC1=CC=C(F)C=C1 XWHHYOYVRVGJJY-UHFFFAOYSA-N 0.000 description 1
- 101710131943 40S ribosomal protein S3a Proteins 0.000 description 1
- IDLISIVVYLGCKO-UHFFFAOYSA-N 6-carboxy-4',5'-dichloro-2',7'-dimethoxyfluorescein Chemical compound O1C(=O)C2=CC=C(C(O)=O)C=C2C21C1=CC(OC)=C(O)C(Cl)=C1OC1=C2C=C(OC)C(O)=C1Cl IDLISIVVYLGCKO-UHFFFAOYSA-N 0.000 description 1
- VWOLRKMFAJUZGM-UHFFFAOYSA-N 6-carboxyrhodamine 6G Chemical compound [Cl-].C=12C=C(C)C(NCC)=CC2=[O+]C=2C=C(NCC)C(C)=CC=2C=1C1=CC(C(O)=O)=CC=C1C(=O)OCC VWOLRKMFAJUZGM-UHFFFAOYSA-N 0.000 description 1
- UKLNSYRWDXRTER-UHFFFAOYSA-N 7-isocyanato-3-phenylchromen-2-one Chemical compound O=C1OC2=CC(N=C=O)=CC=C2C=C1C1=CC=CC=C1 UKLNSYRWDXRTER-UHFFFAOYSA-N 0.000 description 1
- NLSUMBWPPJUVST-UHFFFAOYSA-N 9-isothiocyanatoacridine Chemical compound C1=CC=C2C(N=C=S)=C(C=CC=C3)C3=NC2=C1 NLSUMBWPPJUVST-UHFFFAOYSA-N 0.000 description 1
- ZHQQRIUYLMXDPP-SSDOTTSWSA-N Actinidine Natural products C1=NC=C(C)C2=C1[C@H](C)CC2 ZHQQRIUYLMXDPP-SSDOTTSWSA-N 0.000 description 1
- 102100029457 Adenine phosphoribosyltransferase Human genes 0.000 description 1
- 108010024223 Adenine phosphoribosyltransferase Proteins 0.000 description 1
- 229920000936 Agarose Polymers 0.000 description 1
- OYJCVIGKMXUVKB-GARJFASQSA-N Ala-Leu-Pro Chemical compound C[C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N1CCC[C@@H]1C(=O)O)N OYJCVIGKMXUVKB-GARJFASQSA-N 0.000 description 1
- 108010025188 Alcohol oxidase Proteins 0.000 description 1
- 239000012103 Alexa Fluor 488 Substances 0.000 description 1
- 239000012109 Alexa Fluor 568 Substances 0.000 description 1
- 239000012110 Alexa Fluor 594 Substances 0.000 description 1
- 239000012114 Alexa Fluor 647 Substances 0.000 description 1
- 239000012116 Alexa Fluor 680 Substances 0.000 description 1
- 239000012118 Alexa Fluor 750 Substances 0.000 description 1
- 102100022749 Aminopeptidase N Human genes 0.000 description 1
- 108090000915 Aminopeptidases Proteins 0.000 description 1
- 102000004400 Aminopeptidases Human genes 0.000 description 1
- 108010001779 Ancrod Proteins 0.000 description 1
- 235000002198 Annona diversifolia Nutrition 0.000 description 1
- 108700031308 Antennapedia Homeodomain Proteins 0.000 description 1
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 1
- BHSYMWWMVRPCPA-CYDGBPFRSA-N Arg-Arg-Ile Chemical compound CC[C@H](C)[C@@H](C(O)=O)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@@H](N)CCCN=C(N)N BHSYMWWMVRPCPA-CYDGBPFRSA-N 0.000 description 1
- PTVGLOCPAVYPFG-CIUDSAMLSA-N Arg-Gln-Asp Chemical compound [H]N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(O)=O)C(O)=O PTVGLOCPAVYPFG-CIUDSAMLSA-N 0.000 description 1
- 102220615618 Arginine-tRNA ligase, cytoplasmic_R135G_mutation Human genes 0.000 description 1
- PTNFNTOBUDWHNZ-GUBZILKMSA-N Asn-Arg-Met Chemical compound [H]N[C@@H](CC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCSC)C(O)=O PTNFNTOBUDWHNZ-GUBZILKMSA-N 0.000 description 1
- HCAUEJAQCXVQQM-ACZMJKKPSA-N Asn-Glu-Asp Chemical compound [H]N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(O)=O HCAUEJAQCXVQQM-ACZMJKKPSA-N 0.000 description 1
- KHCNTVRVAYCPQE-CIUDSAMLSA-N Asn-Lys-Asn Chemical compound [H]N[C@@H](CC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(N)=O)C(O)=O KHCNTVRVAYCPQE-CIUDSAMLSA-N 0.000 description 1
- 241000228212 Aspergillus Species 0.000 description 1
- 240000006439 Aspergillus oryzae Species 0.000 description 1
- 235000002247 Aspergillus oryzae Nutrition 0.000 description 1
- 241000228251 Aspergillus phoenicis Species 0.000 description 1
- 241000131386 Aspergillus sojae Species 0.000 description 1
- 108090000658 Astacin Proteins 0.000 description 1
- 102000034498 Astacin Human genes 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 241000714235 Avian retrovirus Species 0.000 description 1
- 241000194108 Bacillus licheniformis Species 0.000 description 1
- 241000194110 Bacillus sp. (in: Bacteria) Species 0.000 description 1
- 108010077805 Bacterial Proteins Proteins 0.000 description 1
- 102100026189 Beta-galactosidase Human genes 0.000 description 1
- 241001446316 Bohle iridovirus Species 0.000 description 1
- 238000009010 Bradford assay Methods 0.000 description 1
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 1
- 108010049990 CD13 Antigens Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282832 Camelidae Species 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 101710167800 Capsid assembly scaffolding protein Proteins 0.000 description 1
- 101710132601 Capsid protein Proteins 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 108090000087 Carboxypeptidase B Proteins 0.000 description 1
- 102000003670 Carboxypeptidase B Human genes 0.000 description 1
- 108010006303 Carboxypeptidases Proteins 0.000 description 1
- 102000005367 Carboxypeptidases Human genes 0.000 description 1
- 108010080937 Carboxypeptidases A Proteins 0.000 description 1
- 102000000496 Carboxypeptidases A Human genes 0.000 description 1
- 108090000397 Caspase 3 Proteins 0.000 description 1
- 108090000425 Caspase 6 Proteins 0.000 description 1
- 102000004018 Caspase 6 Human genes 0.000 description 1
- 108090000567 Caspase 7 Proteins 0.000 description 1
- 102100035904 Caspase-1 Human genes 0.000 description 1
- 102100026549 Caspase-10 Human genes 0.000 description 1
- 108090000572 Caspase-10 Proteins 0.000 description 1
- 102000004066 Caspase-12 Human genes 0.000 description 1
- 108090000570 Caspase-12 Proteins 0.000 description 1
- 102100032616 Caspase-2 Human genes 0.000 description 1
- 108090000552 Caspase-2 Proteins 0.000 description 1
- 102100029855 Caspase-3 Human genes 0.000 description 1
- 102100025597 Caspase-4 Human genes 0.000 description 1
- 101710090338 Caspase-4 Proteins 0.000 description 1
- 102100038916 Caspase-5 Human genes 0.000 description 1
- 101710090333 Caspase-5 Proteins 0.000 description 1
- 102100038902 Caspase-7 Human genes 0.000 description 1
- 102100026548 Caspase-8 Human genes 0.000 description 1
- 108090000538 Caspase-8 Proteins 0.000 description 1
- 102100026550 Caspase-9 Human genes 0.000 description 1
- 108090000566 Caspase-9 Proteins 0.000 description 1
- 102000005572 Cathepsin A Human genes 0.000 description 1
- 108010059081 Cathepsin A Proteins 0.000 description 1
- 108090000712 Cathepsin B Proteins 0.000 description 1
- 102000004225 Cathepsin B Human genes 0.000 description 1
- 102000003902 Cathepsin C Human genes 0.000 description 1
- 108090000267 Cathepsin C Proteins 0.000 description 1
- 102000003908 Cathepsin D Human genes 0.000 description 1
- 108090000258 Cathepsin D Proteins 0.000 description 1
- 102000004178 Cathepsin E Human genes 0.000 description 1
- 108090000611 Cathepsin E Proteins 0.000 description 1
- 102000004173 Cathepsin G Human genes 0.000 description 1
- 108090000617 Cathepsin G Proteins 0.000 description 1
- 108090000619 Cathepsin H Proteins 0.000 description 1
- 102000004175 Cathepsin H Human genes 0.000 description 1
- 108090000624 Cathepsin L Proteins 0.000 description 1
- 102000004172 Cathepsin L Human genes 0.000 description 1
- 241000701489 Cauliflower mosaic virus Species 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- 241001529572 Chaceon affinis Species 0.000 description 1
- 241000282552 Chlorocebus aethiops Species 0.000 description 1
- 239000005496 Chlorsulfuron Substances 0.000 description 1
- VYZAMTAEIAYCRO-UHFFFAOYSA-N Chromium Chemical compound [Cr] VYZAMTAEIAYCRO-UHFFFAOYSA-N 0.000 description 1
- 102100024539 Chymase Human genes 0.000 description 1
- 108090000227 Chymases Proteins 0.000 description 1
- 108090001069 Chymopapain Proteins 0.000 description 1
- 101710094648 Coat protein Proteins 0.000 description 1
- 108700010070 Codon Usage Proteins 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 108010028774 Complement C1 Proteins 0.000 description 1
- 108010078044 Complement C1r Proteins 0.000 description 1
- 102100030149 Complement C1r subcomponent Human genes 0.000 description 1
- 102100025406 Complement C1s subcomponent Human genes 0.000 description 1
- 108090000044 Complement Factor I Proteins 0.000 description 1
- 102000003706 Complement factor D Human genes 0.000 description 1
- 108090000059 Complement factor D Proteins 0.000 description 1
- 102100035431 Complement factor I Human genes 0.000 description 1
- 108091029430 CpG site Proteins 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- MIKUYHXYGGJMLM-GIMIYPNGSA-N Crotonoside Natural products C1=NC2=C(N)NC(=O)N=C2N1[C@H]1O[C@@H](CO)[C@H](O)[C@@H]1O MIKUYHXYGGJMLM-GIMIYPNGSA-N 0.000 description 1
- 241000252867 Cupriavidus metallidurans Species 0.000 description 1
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 1
- NYHBQMYGNKIUIF-UHFFFAOYSA-N D-guanosine Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(CO)C(O)C1O NYHBQMYGNKIUIF-UHFFFAOYSA-N 0.000 description 1
- 101150074155 DHFR gene Proteins 0.000 description 1
- 108091008102 DNA aptamers Proteins 0.000 description 1
- 239000003155 DNA primer Substances 0.000 description 1
- 238000001712 DNA sequencing Methods 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- 102000016928 DNA-directed DNA polymerase Human genes 0.000 description 1
- 108010014303 DNA-directed DNA polymerase Proteins 0.000 description 1
- 101100239628 Danio rerio myca gene Proteins 0.000 description 1
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- FMKGDHLSXFDSOU-BDPUVYQTSA-N Dienon-Astacin Natural products CC(=C/C=C/C=C(C)/C=C/C=C(C)/C=C/C1=C(C)C(=O)C(=CC1(C)C)O)C=CC=C(/C)C=CC2=C(C)C(=O)C(=CC2(C)C)O FMKGDHLSXFDSOU-BDPUVYQTSA-N 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- 102100025012 Dipeptidyl peptidase 4 Human genes 0.000 description 1
- 206010013883 Dwarfism Diseases 0.000 description 1
- 108700033921 EC 3.4.23.20 Proteins 0.000 description 1
- 208000017701 Endocrine disease Diseases 0.000 description 1
- 102100031780 Endonuclease Human genes 0.000 description 1
- 108010067770 Endopeptidase K Proteins 0.000 description 1
- 108010092208 Endothia aspartic proteinase Proteins 0.000 description 1
- 102100029727 Enteropeptidase Human genes 0.000 description 1
- 108010013369 Enteropeptidase Proteins 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- 229910052693 Europium Inorganic materials 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 108050001049 Extracellular proteins Proteins 0.000 description 1
- 102100027285 Fanconi anemia group B protein Human genes 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 108010088842 Fibrinolysin Proteins 0.000 description 1
- 102100037362 Fibronectin Human genes 0.000 description 1
- 102000002090 Fibronectin type III Human genes 0.000 description 1
- 108050009401 Fibronectin type III Proteins 0.000 description 1
- 108010067306 Fibronectins Proteins 0.000 description 1
- 108090000270 Ficain Proteins 0.000 description 1
- 229920001917 Ficoll Polymers 0.000 description 1
- 241000192125 Firmicutes Species 0.000 description 1
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 1
- 238000005033 Fourier transform infrared spectroscopy Methods 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 108090001126 Furin Proteins 0.000 description 1
- 102100035233 Furin Human genes 0.000 description 1
- 101000930822 Giardia intestinalis Dipeptidyl-peptidase 4 Proteins 0.000 description 1
- NUSWUSKZRCGFEX-FXQIFTODSA-N Glu-Glu-Cys Chemical compound OC(=O)CC[C@H](N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CS)C(O)=O NUSWUSKZRCGFEX-FXQIFTODSA-N 0.000 description 1
- 102000053187 Glucuronidase Human genes 0.000 description 1
- 108010060309 Glucuronidase Proteins 0.000 description 1
- 108010051815 Glutamyl endopeptidase Proteins 0.000 description 1
- YWAQATDNEKZFFK-BYPYZUCNSA-N Gly-Gly-Ser Chemical compound NCC(=O)NCC(=O)N[C@@H](CO)C(O)=O YWAQATDNEKZFFK-BYPYZUCNSA-N 0.000 description 1
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 1
- 229930186217 Glycolipid Natural products 0.000 description 1
- 102100021181 Golgi phosphoprotein 3 Human genes 0.000 description 1
- 108010010369 HIV Protease Proteins 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 206010019663 Hepatic failure Diseases 0.000 description 1
- 102000009331 Homeodomain Proteins Human genes 0.000 description 1
- 108010048671 Homeodomain Proteins Proteins 0.000 description 1
- 101000914679 Homo sapiens Fanconi anemia group B protein Proteins 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- 229920001612 Hydroxyethyl starch Polymers 0.000 description 1
- LCWXJXMHJVIJFK-UHFFFAOYSA-N Hydroxylysine Natural products NCC(O)CC(N)CC(O)=O LCWXJXMHJVIJFK-UHFFFAOYSA-N 0.000 description 1
- PMMYEEVYMWASQN-DMTCNVIQSA-N Hydroxyproline Chemical compound O[C@H]1CN[C@H](C(O)=O)C1 PMMYEEVYMWASQN-DMTCNVIQSA-N 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 229910000673 Indium arsenide Inorganic materials 0.000 description 1
- GPXJNWSHGFTCBW-UHFFFAOYSA-N Indium phosphide Chemical compound [In]#P GPXJNWSHGFTCBW-UHFFFAOYSA-N 0.000 description 1
- 108020005350 Initiator Codon Proteins 0.000 description 1
- SNDPXSYFESPGGJ-BYPYZUCNSA-N L-2-aminopentanoic acid Chemical compound CCC[C@H](N)C(O)=O SNDPXSYFESPGGJ-BYPYZUCNSA-N 0.000 description 1
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-N L-arginine Chemical compound OC(=O)[C@@H](N)CCCN=C(N)N ODKSFYDXXFIFQN-BYPYZUCNSA-N 0.000 description 1
- RHGKLRLOHDJJDR-BYPYZUCNSA-N L-citrulline Chemical compound NC(=O)NCCC[C@H]([NH3+])C([O-])=O RHGKLRLOHDJJDR-BYPYZUCNSA-N 0.000 description 1
- GGLZPLKKBSSKCX-YFKPBYRVSA-N L-ethionine Chemical compound CCSCC[C@H](N)C(O)=O GGLZPLKKBSSKCX-YFKPBYRVSA-N 0.000 description 1
- FFFHZYDWPBMWHY-VKHMYHEASA-N L-homocysteine Chemical compound OC(=O)[C@@H](N)CCS FFFHZYDWPBMWHY-VKHMYHEASA-N 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- SNDPXSYFESPGGJ-UHFFFAOYSA-N L-norVal-OH Natural products CCCC(N)C(O)=O SNDPXSYFESPGGJ-UHFFFAOYSA-N 0.000 description 1
- LRQKBLKVPFOOQJ-YFKPBYRVSA-N L-norleucine Chemical compound CCCC[C@H]([NH3+])C([O-])=O LRQKBLKVPFOOQJ-YFKPBYRVSA-N 0.000 description 1
- TYYLDKGBCJGJGW-UHFFFAOYSA-N L-tryptophan-L-tyrosine Natural products C=1NC2=CC=CC=C2C=1CC(N)C(=O)NC(C(O)=O)CC1=CC=C(O)C=C1 TYYLDKGBCJGJGW-UHFFFAOYSA-N 0.000 description 1
- 125000000510 L-tryptophano group Chemical group [H]C1=C([H])C([H])=C2N([H])C([H])=C(C([H])([H])[C@@]([H])(C(O[H])=O)N([H])[*])C2=C1[H] 0.000 description 1
- 241000282838 Lama Species 0.000 description 1
- 102000019298 Lipocalin Human genes 0.000 description 1
- 108050006654 Lipocalin Proteins 0.000 description 1
- 241000186779 Listeria monocytogenes Species 0.000 description 1
- 108050004171 Lon proteases Proteins 0.000 description 1
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 1
- AAORVPFVUIHEAB-YUMQZZPRSA-N Lys-Asp-Gly Chemical compound [H]N[C@@H](CCCCN)C(=O)N[C@@H](CC(O)=O)C(=O)NCC(O)=O AAORVPFVUIHEAB-YUMQZZPRSA-N 0.000 description 1
- 108010053229 Lysyl endopeptidase Proteins 0.000 description 1
- 101710125418 Major capsid protein Proteins 0.000 description 1
- GUBGYTABKSRVRQ-PICCSMPSSA-N Maltose Natural products O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@@H](CO)OC(O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-PICCSMPSSA-N 0.000 description 1
- 101100261636 Methanothermobacter marburgensis (strain ATCC BAA-927 / DSM 2133 / JCM 14651 / NBRC 100331 / OCM 82 / Marburg) trpB2 gene Proteins 0.000 description 1
- 101710181812 Methionine aminopeptidase Proteins 0.000 description 1
- 241000204795 Muraena helena Species 0.000 description 1
- 101100479036 Mus musculus Aars1 gene Proteins 0.000 description 1
- 101000933115 Mus musculus Caspase-4 Proteins 0.000 description 1
- 208000021642 Muscular disease Diseases 0.000 description 1
- KWYHDKDOAIKMQN-UHFFFAOYSA-N N,N,N',N'-tetramethylethylenediamine Chemical compound CN(C)CCN(C)C KWYHDKDOAIKMQN-UHFFFAOYSA-N 0.000 description 1
- NQTADLQHYWFPDB-UHFFFAOYSA-N N-Hydroxysuccinimide Chemical class ON1C(=O)CCC1=O NQTADLQHYWFPDB-UHFFFAOYSA-N 0.000 description 1
- 230000004988 N-glycosylation Effects 0.000 description 1
- KZNQNBZMBZJQJO-UHFFFAOYSA-N N-glycyl-L-proline Natural products NCC(=O)N1CCCC1C(O)=O KZNQNBZMBZJQJO-UHFFFAOYSA-N 0.000 description 1
- 125000001429 N-terminal alpha-amino-acid group Chemical group 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- RHGKLRLOHDJJDR-UHFFFAOYSA-N Ndelta-carbamoyl-DL-ornithine Natural products OC(=O)C(N)CCCNC(N)=O RHGKLRLOHDJJDR-UHFFFAOYSA-N 0.000 description 1
- 229930193140 Neomycin Natural products 0.000 description 1
- 108090000028 Neprilysin Proteins 0.000 description 1
- 102100028782 Neprilysin Human genes 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 101710163270 Nuclease Proteins 0.000 description 1
- 108091005461 Nucleic proteins Chemical group 0.000 description 1
- 101710141454 Nucleoprotein Proteins 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 108020005187 Oligonucleotide Probes Proteins 0.000 description 1
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 1
- UTJLXEIPEHZYQJ-UHFFFAOYSA-N Ornithine Natural products OC(=O)C(C)CCCN UTJLXEIPEHZYQJ-UHFFFAOYSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000194105 Paenibacillus polymyxa Species 0.000 description 1
- 102000007079 Peptide Fragments Human genes 0.000 description 1
- 108010033276 Peptide Fragments Proteins 0.000 description 1
- 108090000882 Peptidyl-Dipeptidase A Proteins 0.000 description 1
- 102000004270 Peptidyl-Dipeptidase A Human genes 0.000 description 1
- 108010030544 Peptidyl-Lys metalloendopeptidase Proteins 0.000 description 1
- WEMYTDDMDBLPMI-DKIMLUQUSA-N Phe-Ile-Lys Chemical compound CC[C@H](C)[C@@H](C(=O)N[C@@H](CCCCN)C(=O)O)NC(=O)[C@H](CC1=CC=CC=C1)N WEMYTDDMDBLPMI-DKIMLUQUSA-N 0.000 description 1
- MSHZERMPZKCODG-ACRUOGEOSA-N Phe-Leu-Phe Chemical compound C([C@H](N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)C1=CC=CC=C1 MSHZERMPZKCODG-ACRUOGEOSA-N 0.000 description 1
- 241001662443 Phemeranthus parviflorus Species 0.000 description 1
- 108700019535 Phosphoprotein Phosphatases Proteins 0.000 description 1
- 102000045595 Phosphoprotein Phosphatases Human genes 0.000 description 1
- 101100124346 Photorhabdus laumondii subsp. laumondii (strain DSM 15139 / CIP 105565 / TT01) hisCD gene Proteins 0.000 description 1
- 241000235648 Pichia Species 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 108010039918 Polylysine Proteins 0.000 description 1
- SVXXJYJCRNKDDE-AVGNSLFASA-N Pro-Pro-His Chemical compound C([C@@H](C(=O)O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H]1NCCC1)C1=CN=CN1 SVXXJYJCRNKDDE-AVGNSLFASA-N 0.000 description 1
- 101710130420 Probable capsid assembly scaffolding protein Proteins 0.000 description 1
- 101710083689 Probable capsid protein Proteins 0.000 description 1
- 108010059712 Pronase Proteins 0.000 description 1
- 108010072866 Prostate-Specific Antigen Proteins 0.000 description 1
- 102100038358 Prostate-specific antigen Human genes 0.000 description 1
- 101800001753 Protease Proteins 0.000 description 1
- 101800004937 Protein C Proteins 0.000 description 1
- 102000017975 Protein C Human genes 0.000 description 1
- 101710149951 Protein Tat Proteins 0.000 description 1
- 108010026552 Proteome Proteins 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 108090000919 Pyroglutamyl-Peptidase I Proteins 0.000 description 1
- 108020004518 RNA Probes Proteins 0.000 description 1
- 108091008103 RNA aptamers Proteins 0.000 description 1
- 239000003391 RNA probe Substances 0.000 description 1
- 230000004570 RNA-binding Effects 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 108091030071 RNAI Proteins 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 241000700157 Rattus norvegicus Species 0.000 description 1
- 101001091368 Rattus norvegicus Glandular kallikrein-7, submandibular/renal Proteins 0.000 description 1
- 208000001647 Renal Insufficiency Diseases 0.000 description 1
- 108090000783 Renin Proteins 0.000 description 1
- 102100028255 Renin Human genes 0.000 description 1
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 description 1
- 241000952054 Rhizopus sp. Species 0.000 description 1
- 108010003581 Ribulose-bisphosphate carboxylase Proteins 0.000 description 1
- 101000898773 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) Saccharopepsin Proteins 0.000 description 1
- 101800001700 Saposin-D Proteins 0.000 description 1
- 101710204410 Scaffold protein Proteins 0.000 description 1
- 241000242678 Schistosoma Species 0.000 description 1
- QMCDMHWAKMUGJE-IHRRRGAJSA-N Ser-Phe-Val Chemical compound [H]N[C@@H](CO)C(=O)N[C@@H](CC1=CC=CC=C1)C(=O)N[C@@H](C(C)C)C(O)=O QMCDMHWAKMUGJE-IHRRRGAJSA-N 0.000 description 1
- FZXOPYUEQGDGMS-ACZMJKKPSA-N Ser-Ser-Gln Chemical compound [H]N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(N)=O)C(O)=O FZXOPYUEQGDGMS-ACZMJKKPSA-N 0.000 description 1
- DKGRNFUXVTYRAS-UBHSHLNASA-N Ser-Ser-Trp Chemical compound [H]N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC1=CNC2=C1C=CC=C2)C(O)=O DKGRNFUXVTYRAS-UBHSHLNASA-N 0.000 description 1
- 108090000899 Serralysin Proteins 0.000 description 1
- 108010034546 Serratia marcescens nuclease Proteins 0.000 description 1
- XUIMIQQOPSSXEZ-UHFFFAOYSA-N Silicon Chemical compound [Si] XUIMIQQOPSSXEZ-UHFFFAOYSA-N 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 229930182558 Sterol Natural products 0.000 description 1
- PJANXHGTPQOBST-VAWYXSNFSA-N Stilbene Natural products C=1C=CC=CC=1/C=C/C1=CC=CC=C1 PJANXHGTPQOBST-VAWYXSNFSA-N 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 108010023197 Streptokinase Proteins 0.000 description 1
- 241000187392 Streptomyces griseus Species 0.000 description 1
- 108010056079 Subtilisins Proteins 0.000 description 1
- 102000005158 Subtilisins Human genes 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 230000006044 T cell activation Effects 0.000 description 1
- 101710137500 T7 RNA polymerase Proteins 0.000 description 1
- 108091046869 Telomeric non-coding RNA Proteins 0.000 description 1
- 102000006601 Thymidine Kinase Human genes 0.000 description 1
- 108020004440 Thymidine kinase Proteins 0.000 description 1
- ISWQCIVKKSOKNN-UHFFFAOYSA-L Tiron Chemical compound [Na+].[Na+].OC1=CC(S([O-])(=O)=O)=CC(S([O-])(=O)=O)=C1O ISWQCIVKKSOKNN-UHFFFAOYSA-L 0.000 description 1
- 108090000373 Tissue Plasminogen Activator Proteins 0.000 description 1
- 102000003978 Tissue Plasminogen Activator Human genes 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000004338 Transferrin Human genes 0.000 description 1
- 108090000901 Transferrin Proteins 0.000 description 1
- 241000223104 Trypanosoma Species 0.000 description 1
- 102000001400 Tryptase Human genes 0.000 description 1
- 108060005989 Tryptase Proteins 0.000 description 1
- RMRFSFXLFWWAJZ-HJOGWXRNSA-N Tyr-Tyr-Tyr Chemical compound C([C@H](N)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(O)=O)C1=CC=C(O)C=C1 RMRFSFXLFWWAJZ-HJOGWXRNSA-N 0.000 description 1
- 108090000848 Ubiquitin Proteins 0.000 description 1
- 102000044159 Ubiquitin Human genes 0.000 description 1
- 101710100170 Unknown protein Proteins 0.000 description 1
- 102000003990 Urokinase-type plasminogen activator Human genes 0.000 description 1
- 108090000435 Urokinase-type plasminogen activator Proteins 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- 244000000188 Vaccinium ovalifolium Species 0.000 description 1
- PAPWZOJOLKZEFR-AVGNSLFASA-N Val-Arg-Lys Chemical compound CC(C)[C@@H](C(=O)N[C@@H](CCCN=C(N)N)C(=O)N[C@@H](CCCCN)C(=O)O)N PAPWZOJOLKZEFR-AVGNSLFASA-N 0.000 description 1
- ZVNYJIZDIRKMBF-UHFFFAOYSA-N Vesnarinone Chemical compound C1=C(OC)C(OC)=CC=C1C(=O)N1CCN(C=2C=C3CCC(=O)NC3=CC=2)CC1 ZVNYJIZDIRKMBF-UHFFFAOYSA-N 0.000 description 1
- IXKSXJFAGXLQOQ-XISFHERQSA-N WHWLQLKPGQPMY Chemical compound C([C@@H](C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N1CCC[C@H]1C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(O)=O)NC(=O)[C@@H](N)CC=1C2=CC=CC=C2NC=1)C1=CNC=N1 IXKSXJFAGXLQOQ-XISFHERQSA-N 0.000 description 1
- 102100039662 Xaa-Pro dipeptidase Human genes 0.000 description 1
- 108020002494 acetyltransferase Proteins 0.000 description 1
- 102000005421 acetyltransferase Human genes 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- DPKHZNPWBDQZCN-UHFFFAOYSA-N acridine orange free base Chemical compound C1=CC(N(C)C)=CC2=NC3=CC(N(C)C)=CC=C3C=C21 DPKHZNPWBDQZCN-UHFFFAOYSA-N 0.000 description 1
- 108090000350 actinidain Proteins 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000010933 acylation Effects 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 229960005305 adenosine Drugs 0.000 description 1
- 150000003838 adenosines Chemical class 0.000 description 1
- 230000011759 adipose tissue development Effects 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 125000000266 alpha-aminoacyl group Chemical group 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229940126575 aminoglycoside Drugs 0.000 description 1
- 229960004233 ancrod Drugs 0.000 description 1
- 208000007502 anemia Diseases 0.000 description 1
- 125000000129 anionic group Chemical group 0.000 description 1
- 239000004410 anthocyanin Substances 0.000 description 1
- 235000010208 anthocyanin Nutrition 0.000 description 1
- 229930002877 anthocyanin Natural products 0.000 description 1
- 150000004636 anthocyanins Chemical class 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 229940009098 aspartate Drugs 0.000 description 1
- 108010038633 aspartylglutamate Proteins 0.000 description 1
- 235000003676 astacin Nutrition 0.000 description 1
- 210000001130 astrocyte Anatomy 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 210000003651 basophil Anatomy 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 229940049706 benzodiazepine Drugs 0.000 description 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- DRTQHJPVMGBUCF-PSQAKQOGSA-N beta-L-uridine Natural products O[C@H]1[C@@H](O)[C@H](CO)O[C@@H]1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-PSQAKQOGSA-N 0.000 description 1
- 125000003164 beta-aspartyl group Chemical group 0.000 description 1
- GUBGYTABKSRVRQ-QUYVBRFLSA-N beta-maltose Chemical compound OC[C@H]1O[C@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@@H]1O GUBGYTABKSRVRQ-QUYVBRFLSA-N 0.000 description 1
- 230000000975 bioactive effect Effects 0.000 description 1
- 239000003124 biologic agent Substances 0.000 description 1
- 239000012472 biological sample Substances 0.000 description 1
- 229920001222 biopolymer Polymers 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 230000036772 blood pressure Effects 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 239000010839 body fluid Substances 0.000 description 1
- 230000036760 body temperature Effects 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- UHYPYGJEEGLRJD-UHFFFAOYSA-N cadmium(2+);selenium(2-) Chemical compound [Se-2].[Cd+2] UHYPYGJEEGLRJD-UHFFFAOYSA-N 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 125000004432 carbon atom Chemical group C* 0.000 description 1
- 150000004649 carbonic acid derivatives Chemical class 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 230000021523 carboxylation Effects 0.000 description 1
- 238000006473 carboxylation reaction Methods 0.000 description 1
- 150000001735 carboxylic acids Chemical class 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 108010054847 carboxypeptidase P Proteins 0.000 description 1
- 108010018550 caspase 13 Proteins 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 125000002091 cationic group Chemical group 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000006037 cell lysis Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000000919 ceramic Substances 0.000 description 1
- 208000019065 cervical carcinoma Diseases 0.000 description 1
- 239000013043 chemical agent Substances 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- VJYIFXVZLXQVHO-UHFFFAOYSA-N chlorsulfuron Chemical compound COC1=NC(C)=NC(NC(=O)NS(=O)(=O)C=2C(=CC=CC=2)Cl)=N1 VJYIFXVZLXQVHO-UHFFFAOYSA-N 0.000 description 1
- 229910052804 chromium Inorganic materials 0.000 description 1
- 239000011651 chromium Substances 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 229960002976 chymopapain Drugs 0.000 description 1
- 229940080701 chymosin Drugs 0.000 description 1
- 238000002983 circular dichroism Methods 0.000 description 1
- 229960002173 citrulline Drugs 0.000 description 1
- 235000013477 citrulline Nutrition 0.000 description 1
- 108090001092 clostripain Proteins 0.000 description 1
- 229910017052 cobalt Inorganic materials 0.000 description 1
- 239000010941 cobalt Substances 0.000 description 1
- GUTLYIVDDKVIGB-UHFFFAOYSA-N cobalt atom Chemical compound [Co] GUTLYIVDDKVIGB-UHFFFAOYSA-N 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 238000002742 combinatorial mutagenesis Methods 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 239000012141 concentrate Substances 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000012926 crystallographic analysis Methods 0.000 description 1
- 108090000200 cucumisin Proteins 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 150000001945 cysteines Chemical class 0.000 description 1
- 229940104302 cytosine Drugs 0.000 description 1
- 210000000172 cytosol Anatomy 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 239000007857 degradation product Substances 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- YSMODUONRAFBET-UHFFFAOYSA-N delta-DL-hydroxylysine Natural products NCC(O)CCC(N)C(O)=O YSMODUONRAFBET-UHFFFAOYSA-N 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 238000011082 depyrogenation Methods 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- MXHRCPNRJAMMIM-UHFFFAOYSA-N desoxyuridine Natural products C1C(O)C(CO)OC1N1C(=O)NC(=O)C=C1 MXHRCPNRJAMMIM-UHFFFAOYSA-N 0.000 description 1
- 229910052805 deuterium Inorganic materials 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 238000002405 diagnostic procedure Methods 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 238000000113 differential scanning calorimetry Methods 0.000 description 1
- 102000038379 digestive enzymes Human genes 0.000 description 1
- 108091007734 digestive enzymes Proteins 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-J diphosphate(4-) Chemical compound [O-]P([O-])(=O)OP([O-])([O-])=O XPPKVPWEQAFLFU-UHFFFAOYSA-J 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- 208000016097 disease of metabolism Diseases 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- PMMYEEVYMWASQN-UHFFFAOYSA-N dl-hydroxyproline Natural products OC1C[NH2+]C(C([O-])=O)C1 PMMYEEVYMWASQN-UHFFFAOYSA-N 0.000 description 1
- 238000009510 drug design Methods 0.000 description 1
- 229940126534 drug product Drugs 0.000 description 1
- 238000007877 drug screening Methods 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 108010003914 endoproteinase Asp-N Proteins 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 210000003979 eosinophil Anatomy 0.000 description 1
- 230000008029 eradication Effects 0.000 description 1
- YSMODUONRAFBET-UHNVWZDZSA-N erythro-5-hydroxy-L-lysine Chemical compound NC[C@H](O)CC[C@H](N)C(O)=O YSMODUONRAFBET-UHNVWZDZSA-N 0.000 description 1
- 230000032050 esterification Effects 0.000 description 1
- 238000005886 esterification reaction Methods 0.000 description 1
- YYXLGGIKSIZHSF-UHFFFAOYSA-N ethene;furan-2,5-dione Chemical compound C=C.O=C1OC(=O)C=C1 YYXLGGIKSIZHSF-UHFFFAOYSA-N 0.000 description 1
- VYXSBFYARXAAKO-UHFFFAOYSA-N ethyl 2-[3-(ethylamino)-6-ethylimino-2,7-dimethylxanthen-9-yl]benzoate;hydron;chloride Chemical compound [Cl-].C1=2C=C(C)C(NCC)=CC=2OC2=CC(=[NH+]CC)C(C)=CC2=C1C1=CC=CC=C1C(=O)OCC VYXSBFYARXAAKO-UHFFFAOYSA-N 0.000 description 1
- OGPBJKLSAFTDLK-UHFFFAOYSA-N europium atom Chemical compound [Eu] OGPBJKLSAFTDLK-UHFFFAOYSA-N 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 210000000887 face Anatomy 0.000 description 1
- 238000010265 fast atom bombardment Methods 0.000 description 1
- 238000000855 fermentation Methods 0.000 description 1
- 230000004151 fermentation Effects 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 235000019836 ficin Nutrition 0.000 description 1
- POTUGHMKJGOKRI-UHFFFAOYSA-N ficin Chemical compound FI=CI=N POTUGHMKJGOKRI-UHFFFAOYSA-N 0.000 description 1
- GVEPBJHOBDJJJI-UHFFFAOYSA-N fluoranthrene Natural products C1=CC(C2=CC=CC=C22)=C3C2=CC=CC3=C1 GVEPBJHOBDJJJI-UHFFFAOYSA-N 0.000 description 1
- 108091006047 fluorescent proteins Proteins 0.000 description 1
- 102000034287 fluorescent proteins Human genes 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 238000013467 fragmentation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 230000037433 frameshift Effects 0.000 description 1
- 230000002538 fungal effect Effects 0.000 description 1
- 108010062699 gamma-Glutamyl Hydrolase Proteins 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 102000034356 gene-regulatory proteins Human genes 0.000 description 1
- 108091006104 gene-regulatory proteins Proteins 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 229940049906 glutamate Drugs 0.000 description 1
- 229930195712 glutamate Natural products 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 150000002313 glycerolipids Chemical class 0.000 description 1
- 150000002314 glycerols Polymers 0.000 description 1
- 150000002334 glycols Chemical class 0.000 description 1
- 108010077515 glycylproline Proteins 0.000 description 1
- 229940029575 guanosine Drugs 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 235000015220 hamburgers Nutrition 0.000 description 1
- 230000002489 hematologic effect Effects 0.000 description 1
- 208000018706 hematopoietic system disease Diseases 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 230000002363 herbicidal effect Effects 0.000 description 1
- 239000004009 herbicide Substances 0.000 description 1
- 101150113423 hisD gene Proteins 0.000 description 1
- 150000002411 histidines Chemical class 0.000 description 1
- 239000012456 homogeneous solution Substances 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 108091008147 housekeeping proteins Proteins 0.000 description 1
- 229920001477 hydrophilic polymer Polymers 0.000 description 1
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 1
- QJHBJHUKURJDLG-UHFFFAOYSA-N hydroxy-L-lysine Natural products NCCCCC(NO)C(O)=O QJHBJHUKURJDLG-UHFFFAOYSA-N 0.000 description 1
- 229940050526 hydroxyethylstarch Drugs 0.000 description 1
- 230000033444 hydroxylation Effects 0.000 description 1
- 238000005805 hydroxylation reaction Methods 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 230000007954 hypoxia Effects 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 208000026278 immune system disease Diseases 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000011503 in vivo imaging Methods 0.000 description 1
- 210000003000 inclusion body Anatomy 0.000 description 1
- RPQDHPTXJYYUPQ-UHFFFAOYSA-N indium arsenide Chemical compound [In]#[As] RPQDHPTXJYYUPQ-UHFFFAOYSA-N 0.000 description 1
- PZOUSPYUWWUPPK-UHFFFAOYSA-N indole Natural products CC1=CC=CC2=C1C=CN2 PZOUSPYUWWUPPK-UHFFFAOYSA-N 0.000 description 1
- RKJUIXBNRJVNHR-UHFFFAOYSA-N indolenine Natural products C1=CC=C2CC=NC2=C1 RKJUIXBNRJVNHR-UHFFFAOYSA-N 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000007689 inspection Methods 0.000 description 1
- 239000000543 intermediate Substances 0.000 description 1
- RGXCTRIQQODGIZ-UHFFFAOYSA-O isodesmosine Chemical compound OC(=O)C(N)CCCC[N+]1=CC(CCC(N)C(O)=O)=CC(CCC(N)C(O)=O)=C1CCCC(N)C(O)=O RGXCTRIQQODGIZ-UHFFFAOYSA-O 0.000 description 1
- BPHPUYQFMNQIOC-NXRLNHOXSA-N isopropyl beta-D-thiogalactopyranoside Chemical compound CC(C)S[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O BPHPUYQFMNQIOC-NXRLNHOXSA-N 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- 201000006370 kidney failure Diseases 0.000 description 1
- 101150066555 lacZ gene Proteins 0.000 description 1
- 150000002605 large molecules Chemical class 0.000 description 1
- 239000010410 layer Substances 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 231100000225 lethality Toxicity 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000029226 lipidation Effects 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 238000004811 liquid chromatography Methods 0.000 description 1
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 208000007903 liver failure Diseases 0.000 description 1
- 231100000835 liver failure Toxicity 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 210000005265 lung cell Anatomy 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 229920001427 mPEG Polymers 0.000 description 1
- 239000006249 magnetic particle Substances 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 229910021645 metal ion Inorganic materials 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 108010009355 microbial metalloproteinases Proteins 0.000 description 1
- 230000003228 microsomal effect Effects 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 230000037230 mobility Effects 0.000 description 1
- 108091005601 modified peptides Proteins 0.000 description 1
- 230000004001 molecular interaction Effects 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 238000002887 multiple sequence alignment Methods 0.000 description 1
- 230000003387 muscular Effects 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 210000003643 myeloid progenitor cell Anatomy 0.000 description 1
- GNOLWGAJQVLBSM-UHFFFAOYSA-N n,n,5,7-tetramethyl-1,2,3,4-tetrahydronaphthalen-1-amine Chemical compound C1=C(C)C=C2C(N(C)C)CCCC2=C1C GNOLWGAJQVLBSM-UHFFFAOYSA-N 0.000 description 1
- 210000004897 n-terminal region Anatomy 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 230000001537 neural effect Effects 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 230000000626 neurodegenerative effect Effects 0.000 description 1
- 230000004766 neurogenesis Effects 0.000 description 1
- 201000001119 neuropathy Diseases 0.000 description 1
- 230000007823 neuropathy Effects 0.000 description 1
- 150000002825 nitriles Chemical class 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- IJGRMHOSHXDMSA-UHFFFAOYSA-N nitrogen Substances N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 108091027963 non-coding RNA Proteins 0.000 description 1
- 102000042567 non-coding RNA Human genes 0.000 description 1
- 210000000633 nuclear envelope Anatomy 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 230000009965 odorless effect Effects 0.000 description 1
- 150000002482 oligosaccharides Polymers 0.000 description 1
- 108010003052 omptin outer membrane protease Proteins 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 229960003104 ornithine Drugs 0.000 description 1
- 230000011164 ossification Effects 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 230000003071 parasitic effect Effects 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 238000012510 peptide mapping method Methods 0.000 description 1
- 208000033808 peripheral neuropathy Diseases 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 102000013415 peroxidase activity proteins Human genes 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- WTJKGGKOPKCXLL-RRHRGVEJSA-N phosphatidylcholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCC=CCCCCCCCC WTJKGGKOPKCXLL-RRHRGVEJSA-N 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 230000001817 pituitary effect Effects 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 229940012957 plasmin Drugs 0.000 description 1
- 229920000724 poly(L-arginine) polymer Polymers 0.000 description 1
- 229920000233 poly(alkylene oxides) Polymers 0.000 description 1
- 229920001308 poly(aminoacid) Polymers 0.000 description 1
- 229920001583 poly(oxyethylated polyols) Polymers 0.000 description 1
- 108010011110 polyarginine Proteins 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 238000006116 polymerization reaction Methods 0.000 description 1
- 229920001451 polypropylene glycol Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 210000000229 preadipocyte Anatomy 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 210000004986 primary T-cell Anatomy 0.000 description 1
- 239000012602 primary packaging material Substances 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000009465 prokaryotic expression Effects 0.000 description 1
- 108010066823 proline dipeptidase Proteins 0.000 description 1
- 108010043535 protease S Proteins 0.000 description 1
- 229960000856 protein c Drugs 0.000 description 1
- 108020001580 protein domains Proteins 0.000 description 1
- 238000002818 protein evolution Methods 0.000 description 1
- 238000000751 protein extraction Methods 0.000 description 1
- 238000010377 protein imaging Methods 0.000 description 1
- 239000012460 protein solution Substances 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 108091006091 regulatory enzymes Proteins 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 108090000588 rhizopuspepsin Proteins 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 102220202181 rs11557637 Human genes 0.000 description 1
- 102220202187 rs2290629 Human genes 0.000 description 1
- 102220201842 rs2305727 Human genes 0.000 description 1
- 102220202178 rs2305728 Human genes 0.000 description 1
- 102220203132 rs2305733 Human genes 0.000 description 1
- 102200023405 rs2305734 Human genes 0.000 description 1
- 102220203150 rs2305735 Human genes 0.000 description 1
- 102200023403 rs244903 Human genes 0.000 description 1
- 102220202070 rs34723836 Human genes 0.000 description 1
- 102220405067 rs590784 Human genes 0.000 description 1
- 102220203130 rs61744940 Human genes 0.000 description 1
- 102220203131 rs62385662 Human genes 0.000 description 1
- 102220202075 rs653568 Human genes 0.000 description 1
- 102220301301 rs79948711 Human genes 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 238000007423 screening assay Methods 0.000 description 1
- 125000000467 secondary amino group Chemical group [H]N([*:1])[*:2] 0.000 description 1
- 229930000044 secondary metabolite Natural products 0.000 description 1
- 230000018448 secretion by cell Effects 0.000 description 1
- 238000010187 selection method Methods 0.000 description 1
- 239000006152 selective media Substances 0.000 description 1
- SBIBMFFZSBJNJF-UHFFFAOYSA-N selenium;zinc Chemical compound [Se]=[Zn] SBIBMFFZSBJNJF-UHFFFAOYSA-N 0.000 description 1
- 239000004065 semiconductor Substances 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 238000002864 sequence alignment Methods 0.000 description 1
- 230000000405 serological effect Effects 0.000 description 1
- 210000000717 sertoli cell Anatomy 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 230000037432 silent mutation Effects 0.000 description 1
- 229910052710 silicon Inorganic materials 0.000 description 1
- 239000010703 silicon Substances 0.000 description 1
- 210000002363 skeletal muscle cell Anatomy 0.000 description 1
- 230000022379 skeletal muscle tissue development Effects 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000001509 sodium citrate Substances 0.000 description 1
- NLJMYIDDQXHKNR-UHFFFAOYSA-K sodium citrate Chemical compound O.O.[Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O NLJMYIDDQXHKNR-UHFFFAOYSA-K 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 238000001179 sorption measurement Methods 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 238000004611 spectroscopical analysis Methods 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 230000010473 stable expression Effects 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 150000003432 sterols Chemical class 0.000 description 1
- 235000003702 sterols Nutrition 0.000 description 1
- PJANXHGTPQOBST-UHFFFAOYSA-N stilbene Chemical compound C=1C=CC=CC=1C=CC1=CC=CC=C1 PJANXHGTPQOBST-UHFFFAOYSA-N 0.000 description 1
- 235000021286 stilbenes Nutrition 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 238000003756 stirring Methods 0.000 description 1
- 229960005202 streptokinase Drugs 0.000 description 1
- 230000035882 stress Effects 0.000 description 1
- 108010059339 submandibular proteinase A Proteins 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- BDHFUVZGWQCTTF-UHFFFAOYSA-M sulfonate Chemical compound [O-]S(=O)=O BDHFUVZGWQCTTF-UHFFFAOYSA-M 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000011593 sulfur Substances 0.000 description 1
- 230000008093 supporting effect Effects 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 238000011191 terminal modification Methods 0.000 description 1
- 229940126585 therapeutic drug Drugs 0.000 description 1
- 150000003573 thiols Chemical class 0.000 description 1
- 238000012090 tissue culture technique Methods 0.000 description 1
- 229960000187 tissue plasminogen activator Drugs 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 239000012581 transferrin Substances 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 238000003146 transient transfection Methods 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- 101150081616 trpB gene Proteins 0.000 description 1
- 101150111232 trpB-1 gene Proteins 0.000 description 1
- 229960001322 trypsin Drugs 0.000 description 1
- 108010044292 tryptophyltyrosine Proteins 0.000 description 1
- 238000007039 two-step reaction Methods 0.000 description 1
- 238000000108 ultra-filtration Methods 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 241000701366 unidentified nuclear polyhedrosis viruses Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- DRTQHJPVMGBUCF-UHFFFAOYSA-N uracil arabinoside Natural products OC1C(O)C(CO)OC1N1C(=O)NC(=O)C=C1 DRTQHJPVMGBUCF-UHFFFAOYSA-N 0.000 description 1
- 229940045145 uridine Drugs 0.000 description 1
- 229960005356 urokinase Drugs 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 229920002554 vinyl polymer Polymers 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 239000003643 water by type Substances 0.000 description 1
- 239000001018 xanthene dye Substances 0.000 description 1
- 238000001086 yeast two-hybrid system Methods 0.000 description 1
- 229910052727 yttrium Inorganic materials 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/3955—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/53—Ligases (6)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/14—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
- A61K9/19—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/40—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/10—Transferases (2.)
- C12N9/1085—Transferases (2.) transferring alkyl or aryl groups other than methyl groups (2.5)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/93—Ligases (6)
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/573—Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2121/00—Preparations for use in therapy
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y601/00—Ligases forming carbon-oxygen bonds (6.1)
- C12Y601/01—Ligases forming aminoacyl-tRNA and related compounds (6.1.1)
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/90—Enzymes; Proenzymes
- G01N2333/9015—Ligases (6)
Definitions
- the present invention relates generally to compositions comprising newly identified protein fragments of aminoacyl-tRNA synthetases and other proteins, polynucleotides that encode them and complements thereof, related agents, and methods of use thereof in diagnostic, drug discovery, research, and therapeutic applications.
- AARSs aminoacyl-tRNA synthetases
- these unexpected activities are not observed in the context of the full-length or parental protein sequences; instead, they are observed following removal or resection of AARS protein fragments from their parental sequences, or by expressing and sufficiently purifying fragment AARS sequences and then testing for novel, non-synthetase related activities.
- AARS protein fragments While the full-length sequences of AARS have been known for some time, no systematic experimental analysis has been conducted to elucidate such AARS protein fragments, or protein fragments from related or associated proteins, or to evaluate the potential role of the full length AARS proteins for novel biological activities outside of the context of amino acid synthesis.
- AARS protein fragments, AARS domains, or AARS alternative splice variants are referred to herein as "resectins”.
- resectin refers to a portion of a protein which has been excised or restricted (either by means of proteolysis, alternative splicing, mutagenesis, or recombinant genetic engineering) from the context of its native full-length or parental protein sequence, which often otherwise masks its novel biological activities.
- resectins are not limited to biotherapeutic agents, diagnostic agents, or drug targets in the treatment of various medical conditions, or their potential association with human diseases.
- AARSs As essential housekeeping genes with a known function in mammals that is critical to life, AARSs were neither considered as drug targets in mammals, nor were they parsed out by standard genomic sequencing, bioinformatic, or similar efforts to identify resectins having non-synthetase activities. Standard biochemical research efforts have similarly been directed away from characterizing the biological properties of AARS resectins and their potential therapeutic and diagnostic relevance, mainly due to the previously understood role of their corresponding full-length parental AARSs.
- Embodiments of the present invention relate generally to the discovery of protein fragments of arginyl aminoacyl-tRNA synthetase (ArgRSs), which possess non-canonical biological activities, such as extracellular signaling activities, and/or other characteristics of therapeutic and diagnostic relevance.
- ArgRSs arginyl aminoacyl-tRNA synthetase
- the AARSs are universal and essential elements of the protein synthesis machinery found in all organisms, but human AARSs and their associated proteins have naturally-occurring resected variants, with potent cell signaling activities that contribute to normal functioning of humans.
- the activities of these protein fragments are distinct from the protein synthesis activities commonly known for AARSs, and the present disclosure includes the discovery and development of these resected proteins as new biotherapeutic agents, new discovery research reagents, and as new antigens/targets for directed biologies and diagnostic agents that can be used to potentially treat or diagnose a wide variety of human diseases, such as inflammatory, hematological, neurodegenerative, autoimmune, hematopoietic, cardiovascular, and metabolic diseases or disorders.
- diseases such as inflammatory, hematological, neurodegenerative, autoimmune, hematopoietic, cardiovascular, and metabolic diseases or disorders.
- ArgRS protein fragment(s) of the present disclosure may therefore be referred to as "resectins,” or alternatively as “appendacrines.”
- resectin derives from the process of excising or resecting a given ArgRS protein fragment from the context of its full-length parent ArgRS sequence, which typically masks its non-canonical activities.
- the ArgRS protein fragments and polynucleotides of the present disclosure were identified through the occurrence of this resection process, whether naturally-occurring ( e . g. , proteolytic, splice variant), artificially-induced, or predicted.
- appendacrine derives from a combination of "append” (from Latin - appender) and to “separate” or “discern” (from Greek - crines),” and also reflects the separation of one or more appended domains of the ArgRS protein fragments from their corresponding full-length or parent ArgRS sequences.
- AARS fragments Although a few AARS fragments have been previously shown to have non-synthetase activities, the expression, isolation, purification, and characterization of such fragments for biotherapeutic, discovery, or diagnostic utility is limited, and persons skilled in the art would not have readily appreciated such activities to associate with each member of the entire family of AARSs, or with alternative fragments.
- a methodical approach was utilized to discover and verify AARS protein fragments for the 20 mitochondrial and 20 cytosolic AARSs (and associated proteins) for biotherapeutic discovery and diagnostic utility.
- ArgRS protein fragment(s) and polynucleotides that encode them are identified from biological samples using mass spectrometry (MS), mainly to identify proteolytic fragments, and others were identified by deep sequencing techniques, mainly to identify splice variants.
- MS mass spectrometry
- Other ArgRS protein fragment(s) are identified using in silico predictions of amino acid sequences, such as by computationally comparing synthetases from humans and lower organisms along with key demarcations ( e.g ., protease sites); this approach utilized sequence analysis of the full-length ArgRS based on specific criteria to discern proteolytic fragments and functional domains possessing non-canonical biological activities.
- Novel resectins of the AARSs are unexpected, and their differential expression is also unexpected. Specific resections are typically seen under different treatments (e.g ., from cells grown in media with or without serum), at different stages of growth (e.g., adult brain vs. fetal brain) and for different tissue types (e.g., pancreas vs. liver). The pattern of expression is not the same for all aminoacyl tRNA synthetases despite the fact that the canonical functions for all aminoacyl tRNA synthetases are needed in the same cell locations and in relatively proportional amounts.
- AARS protein fragments can be expressed and purified to sufficiently high purity to discern their biological properties. Previously, fragments were often not of sufficient purity, folding, and stability to enable proper biological characterization of non-synthetase activities. Cell based assays, for instance, are used in conjunction with sufficiently pure, stable, soluble and folded resectins to reveal their important biotherapeutic, discovery or diagnostic activities.
- embodiments of the present disclosure relate to protein fragments of Arginyl tRNA synthetases, related agents and compositions of biotherapeutic, discovery, or diagnostic utility, and methods of use thereof.
- the compositions of the present disclosure are useful in a variety of diagnostic, drug discovery, and therapeutic applications, as described herein.
- the ArgRS proteins and fragments are purified and stored in suitable condition to the extent required for such biotherapeutic, discovery, or diagnostic uses.
- compositions comprising an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least about 100, 90, 80, 70, 60, 50 or 40 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, wherein the protein fragment has an EC 50 of about 10 nM or less for a cell-based non-canonical biological activity and has a solubility of at least about 10 mg/ml, and wherein the composition has a purity of at least about 95% on a protein basis and substantially undetectable levels of endotoxin.
- the composition is substantially serum free.
- assays for measuring such features of resectins are described herein and may be used to define aspects of the invention. In certain aspects, these features will be preferable for biotherapeutic utility of the ArgRS protein fragments described herein.
- compositions comprising an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least about 100, 90, 80, 70, 60, 50 or 40 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, and has a solubility of at least about 5 mg/ml, and wherein the composition has a purity of at least about 95% on a protein basis, and less than about 10 EU / mg protein endotoxin.
- the composition is a therapeutic composition.
- the composition is substantially serum free.
- the ArgRS protein fragment comprises a non-canonical activity.
- the non-canonical biological activity is selected from modulation of extracellular signaling, modulation of cell proliferation, modulation of cell differentiation, modulation of gene transcription, modulation of cytokine production or activity, modulation of cytokine receptor activity, and modulation of inflammation.
- the ArgRS protein fragment has an EC 50 of less than about 1 nM, about 5 nM, about 10 nM, about 50 nM, about 100 nM or about 200 nM for a cell-based non-canonical biological activity.
- the ArgRS protein fragment is fused to a heterologous polypeptide.
- the ArgRS fusion protein substantially retains a non-canonical activity of the ArgRS protein fragment.
- the ArgRS fusion protein suppresses a non-canonical activity of the ArgRS protein fragment.
- the heterologous polypeptide is attached to the N-terminus of the ArgRS protein fragment.
- the heterologous polypeptide is attached to the C-terminus of the ArgRS protein fragment.
- the heterologous polypeptide is selected from the group consisting of purification tags, epitope tags, targeting sequences, signal peptides, membrane translocating sequences, and PK modifiers.
- the composition comprises an ArgRS protein fragment at a concentration of least about 10 mg/mL. In certain embodiments the composition comprises an ArgRS protein fragment which is at least 90% monodisperse. In certain embodiments the composition comprises less than about 3 % high molecular weight aggregated proteins. In certain embodiments the composition exhibits less than 3% aggregation when stored at a concentration of at least 10 mg/ mL in PBS for one week at 4 °C. In certain embodiments the composition exhibits less than 3% aggregation when stored at a concentration of at least 10 mg/ mL in PBS for one week at room temperature.
- compositions comprising an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that differs from an amino acid sequence set forth in SEQ ID NO: 29, 151, or 137 7-9 by substitution, deletion, and/or addition of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids, wherein the altered protein fragment substantially retains a non-canonical activity of the unaltered protein, or has a dominant negative phenotype in relation to the non-canonical activity, wherein the protein fragment has a solubility of at least about 5 mg/ml, and wherein the composition has a purity of at least about 95% on a protein basis and less than about 10 EU / mg protein endotoxin.
- the composition is substantially serum free.
- compositions comprising an isolated antibody that specifically binds to an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment as set forth in SEQ ID NO: 29, 151, or 137, wherein affinity of the antibody for the ArgRS protein fragment is about 10X stronger than its affinity for a corresponding full-length ArgRS polypeptide.
- ArgRS arginyl aminoacyl-tRNA synthetase
- compositions comprising an isolated antibody that specifically binds to an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment as set forth in SEQ ID NO: 29, 151, or 137, wherein the antibody has an affinity of at least about 10 nM for the ArgRS protein fragment, and an affinity of at least about 100 nM for a corresponding full-length ArgRS polypeptide.
- the antibody binds to an epitope located within an ArgRS polypeptide unique splice junction as set forth in any of SEQ ID NO: 29, 151, or 137, or to an amino acid sequence C-terminal of this splice site.
- the antibody antagonizes the non-canonical activity of the ArgRS protein fragment.
- Such antagonists may optionally bind the corresponding parental or full-length ArgRS.
- bioassay systems comprising a substantially pure arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, and a binding partner that binds to the ArgRS protein fragment.
- the binding partner is selected from the group consisting of a cellular surface receptor protein, nucleic acid, lipid membrane, cell regulatory protein, enzyme, and transcription factor.
- a receptor may be part of a cell, preferably a cell relevant to the revealed biology of the resectin.
- Certain embodiments include cellular compositions, comprising an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, and an engineered population of cells in which at least one cell comprises a polynucleotide encoding said ArgRS protein fragment.
- the cells are capable of growing in a serum free medium.
- detection systems comprising a substantially pure arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 50 or 100 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, a cell that comprises a cell-surface receptor or an extracellular portion thereof that binds to the protein fragment, and a molecule of less than about 2000 daltons, or a second polypeptide, which modulates binding or interaction between the ArgRS protein fragment and the extracellular receptor.
- ArgRS arginyl aminoacyl-tRNA synthetase
- Particular embodiments include diagnostic systems, comprising a substantially pure arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, and a cell that comprises a cell-surface receptor or an extracellular portion thereof that binds to the ArgRS protein fragment, wherein the system or cell comprises an indicator molecule that allows detection of a change in the levels or activity of the cell-surface receptor or extracellular portion thereof.
- ArgRS arginyl aminoacyl-tRNA synthetase
- Certain embodiments include cellular growth devices, comprising an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, an engineered population of cells in which at least one cell comprises a polynucleotide encoding said ArgRS protein fragment, at least about 10 liters of serum-free cell media, and a sterile container.
- the cells utilized for any of the methods or compositions described herein are capable of growing in serum-free media, optionally with an antibiotic and an inducer.
- compositions comprising an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, wherein the protein fragment specifically binds to a binding partner and has a solubility of at least about 5 mg/ml, and wherein the composition has a purity of at least about 95% on a protein basis.
- ArgRS arginyl aminoacyl-tRNA synthetase
- the composition may have less than 10 EU endotoxin / mg protein.
- compositions comprising an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that is at least 80%, 85%, 90%, 95%, 98%, or 100% identical to an amino acid sequence set forth in SEQ ID NO: 29, 151, or 137, wherein the protein fragment has a solubility of at least about 5 mg/ml, and wherein the composition has a purity of at least about 95% on a protein basis and less than 10 EU endotoxin / mg protein.
- the compositions may comprise an ArgRS protein fragment that is at least about 50%, about 60%, about 70%, about 80%, about 90% or about 95% monodisperse with respect to its apparent molecular mass.
- compositions comprise less than about 10 % (on a protein basis) high molecular weight aggregated proteins, or less than about 5 % high molecular weight aggregated proteins, or less than about 4% high molecular weight aggregated proteins, or less than about 3% high molecular weight aggregated proteins, or less than 2 % high molecular weight aggregated proteins, or less than about 1% high molecular weight aggregated proteins.
- the compositions exhibits less than about 10% aggregation when stored at a concentration of at least 10 mg/ mL in PBS for one week at 4 °C, or less than about 5% aggregation when stored at a concentration of at least 10 mg/ mL in PBS for one week at 4 °C, or less than about 3% aggregation when stored at a concentration of at least 10 mg/ mL in PBS for one week at 4 °C, or less than about 2% aggregation when stored at a concentration of at least 10 mg/ mL in PBS for one week at 4 °C, or less than about 1% aggregation when stored at a concentration of at least 10 mg/ mL in PBS for one week at 4 °C.
- compositions comprising a substantially pure arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, and at least one covalently or non-covalently moiety attached thereto.
- the moiety is a detectable label.
- the moiety is a water soluble polymer.
- the moiety is PEG.
- the moiety is attached to the N-terminus of the protein fragment.
- the moiety is attached to the C-terminus of the protein fragment.
- compositions comprising a solid substrate attached to an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, or a biologically active fragment or variant thereof, wherein the protein fragment has a solubility of at least about 5 mg/ml, and the composition has a purity of at least about 95% on a protein basis.
- ArgRS arginyl aminoacyl-tRNA synthetase
- compositions comprising a binding agent that specifically binds to an isolated arginyl aminoacyl-tRNA synthetase (ArgRS) protein fragment as set forth in SEQ ID NO: 29, 151, or 137, wherein the binding agent has an affinity of at least about 1 nM for the protein fragment.
- the binding agent binds to an epitope located within an ArgRS polypeptide unique splice junction as set forth in any of SEQ ID NO: 29, 151, or 137, or to an amino acid sequence C-terminal of this splice site.
- the binding agent antagonizes a non-canonical activity of the ArgRS polypeptide.
- Certain embodiments include isolated arginyl aminoacyl-tRNA synthetase (ArgRS) polypeptides, comprising an amino acid sequence of an ArgRS protein fragment as described herein, an amino acid sequence encoded by an ArgRS polynucleotide as described herein, or a variant or fragment thereof.
- Certain ArgRS polypeptides comprise an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, or 100% identical to an ArgRS reference sequence as disclosed in SEQ ID NO: 29, 151, or 137.
- ArgRS polypeptides consist essentially of an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, or 100% identical to an ArgRS reference sequence as disclosed in SEQ ID NO: 29, 151, or 137.
- the polypeptide comprises a non-canonical biological activity.
- the non-canonical biological activity is selected from modulation of cell signaling (e.g ., extracellular signaling), modulation of cell proliferation, modulation of cell migration, modulation of cell differentiation, modulation of apoptosis or cell death, modulation of angiogenesis, modulation of cell binding, modulation of cellular metabolism, modulation of cellular uptake, modulation of gene transcription, or secretion, modulation of cytokine production or activity, modulation of cytokine receptor activity, and modulation of inflammation.
- cell signaling e.g ., extracellular signaling
- antibodies and other binding agents that exhibit binding specificity for an isolated ArgRS polypeptide as described herein, a binding partner of the ArgRS polypeptide, or the complex of both.
- the affinity of the antibody or binding agent for the ArgRS polypeptide is about 10X stronger than its affinity for a corresponding full-length ArgRS polypeptide.
- the binding agent is selected from a peptide, peptide mimetic, an adnectin, an aptamer, and a small molecule.
- the antibody or binding agent antagonizes a non-canonical activity of the ArgRS polypeptide.
- the antibody or binding agent agonizes a non-canonical activity of the ArgRS polypeptide.
- Certain embodiments include isolated arginyl aminoacyl-tRNA synthetase (ArgRS) polynucleotides, comprising a nucleotide sequence of an ArgRS polynucleotide as described herein, a nucleotide sequence that encodes an ArgRS protein fragment as described herein, or a variant, a fragment, or a complement thereof.
- Certain ArgRS polynucleotides comprise a nucleotide sequence that is at least 80%, 85%, 90%, 95%, 98%, or 100% identical to an ArgRS reference polynucleotide, or a complement thereof, as disclosed in SEQ ID NO: 29, 151, or 137.
- the nucleotide sequence is codon optimized for bacterial expression.
- Specific ArgRS polynucleotides consist essentially of a nucleotide sequence that is at least 80%, 85%, 90%, 95%, 98%, or 100% identical to an ArgRS reference polynucleotide, or a complement thereof, as disclosed in SEQ ID NO: 29, 151, or 137.
- Other ArgRS polynucleotides comprise or consist essentially of a nucleotide sequence that specifically hybridizes to an ArgRS reference polynucleotide, as disclosed in SEQ ID NO: 29, 151, or 137.
- the polynucleotide is selected from a primer, a probe, and an antisense oligonucleotide.
- the primer, probe, or antisense oligonucleotide is targeted to a specific or unique splice junction, and / or sequence 3' of this splice site within an ArgRS polynucleotide.
- Certain embodiments include methods of determining presence or levels of an ArgRS protein fragment in a sample, comprising contacting the sample with one or more binding agents that specifically bind to an ArgRS protein fragment as described herein, detecting the presence or absence of the binding agent, and thereby determining the presence or levels of the ArgRS protein fragment.
- Other embodiments include methods of determining presence or levels of an ArgRS protein fragment in a sample, comprising analyzing the sample with a detector that is capable of specifically identifying a protein fragment as described herein, and thereby determining the presence or levels of the ArgRS protein fragment.
- the detector is a mass spectrometer (MS), a flow cytometer, a protein imaging device, an enzyme-linked immunosorbent assays (ELISA), or a protein microarray.
- MS mass spectrometer
- ELISA enzyme-linked immunosorbent assays
- Certain embodiments comprise comparing the presence or levels of the ArgRS protein fragment to a control sample or a predetermined value.
- Certain embodiments comprise characterizing the state of the sample to distinguish it from the control.
- the sample and control comprise a cell or tissue, and the method comprises distinguishing between cells or tissues of different species, cells of different tissues or organs, cells at different cellular developmental states, cells at different cellular differentiation states, cells at different physiological states, or healthy and diseased cells. For instance, selected resectins may be more abundant under conditions such as stress or insult.
- Certain embodiments include discovery methods of, and related compositions for, identifying a compound that specifically binds to an arginyl aminoacyl-tRNA synthetase (ArgRS) polypeptide as described herein, or one or more of its cellular binding partners, comprising a) combining the ArgRS polypeptide or its cellular binding partner or both with at least one test compound under suitable conditions, and b) detecting binding of the ArgRS polypeptide or its cellular binding partner or both to the test compound, thereby identifying a compound that specifically binds to the ArgRS polypeptide or its cellular binding partner or both.
- ArgRS arginyl aminoacyl-tRNA synthetase
- the test compound is a polypeptide or peptide, an antibody or antigen-binding fragment thereof, a peptide mimetic, or a small molecule.
- the test compound agonizes a non-canonical biological activity of the ArgRS polypeptide or its cellular binding partner.
- the test compound antagonizes a non-canonical biological activity of the ArgRS polypeptide or its cellular binding partner.
- Certain embodiments include a compound identified by the above-method, such as an agonist ( e.g ., small molecule, peptide).
- Certain embodiments include methods of determining presence or levels of a polynucleotide sequence of an ArgRS splice variant in a sample, comprising contacting the sample with one or more oligonucleotides that specifically hybridize to an ArgRS polynucleotide as described herein, detecting the presence or absence of the oligonucleotides in the sample, and thereby determining the presence or levels of the polynucleotide sequence of the ArgRS splice variant.
- inventions include methods of determining presence or levels of a polynucleotide sequence of an ArgRS splice variant in a sample, comprising contacting the sample with at least two oligonucleotides that specifically amplify an ArgRS polynucleotide as described herein, performing an amplification reaction, detecting the presence or absence of an amplified product, and thereby determining presence or levels of the polynucleotide sequence of the ArgRS splice variant.
- the oligonucleotide(s) specifically hybridize to or specifically amplify a splice junction that is unique to the ArgRS splice variant.
- Certain embodiments include comparing the presence or levels of the ArgRS protein fragment or splice variant to a control sample or a predetermined value. Certain embodiments include characterizing the state of the sample to distinguish it from the control.
- the sample and control comprise a cell or tissue, and the method comprises distinguishing between cells or tissues of different species, cells of different tissues or organs, cells at different cellular developmental states, cells at different cellular differentiation states, or healthy and diseased cells.
- compositions comprising an ArgRS polynucleotide described herein, an ArgRS polypeptide described herein, a binding agent as described herein, or a compound identified by the above-method or described herein, and a pharmaceutically acceptable excipient or carrier.
- Certain embodiments include methods of modulating a cellular activity of a cell, comprising contacting the cell with an ArgRS polynucleotide described herein, an ArgRS polypeptide described herein, a binding agent described herein, a compound of the above-method or described herein, or a pharmaceutical composition described herein.
- the cellular activity is selected from cell proliferation, cell migration, cell differentiation, apoptosis or cell death, cell signaling, angiogenesis, cell binding, cellular uptake, cell secretion, metabolism, cytokine production or activity, cytokine receptor activity, gene transcription, and inflammation.
- the cell is selected from the group consisting of pre-adipocytes, bone marrow, neutrophils, blood cells, hepatocytes, astrocytes, mesenchymal stem cells, and skeletal muscle cells.
- the cell is in a subject.
- Certain embodiments comprise treating the subject, wherein the subject has a condition associated with a neoplastic disease, an immune system disease or condition, an infectious disease, a metabolic disease, an inflammatory disorder, neuronal/neurological disease, a muscular/cardiovascular disease, a disease associated with aberrant hematopoiesis, a disease associated with aberrant angiogenesis, or a disease associated with aberrant cell survival.
- Also included are processes for manufacturing a pharmaceutical compound comprising: a) performing an in vitro screen of one or more candidate compounds in the presence an ArgRS protein fragment of at least 70 amino acids that comprises an amino acid sequence as set forth in SEQ ID NO: 29, 151, or 137, to identify a compound that specifically binds to the ArgRS protein fragment; b) performing a cell-based or biochemical or receptor assay with the compound identified in step a), to identify a compound that modulates one or more non-canonical activities of the ArgRS protein fragment; c) optionally assessing the structure-activity relationship (SAR) of the compound identified in step b), to correlate its structure with modulation of the non-canonical activity, and optionally derivatizing the compound to alter its ability to modulate the non-canonical activity; and d) producing sufficient amounts of the compound identified in step b), or the derivatized compound in step c), for use in humans, thereby manufacturing the pharmaceutical compound.
- SAR structure-activity relationship
- Other embodiments include processes for manufacturing a pharmaceutical compound, comprising: a) performing an in vitro screen of one or more candidate compounds in the presence a cell-surface receptor or an extracellular portion thereof that specifically binds to an ArgRS protein fragment of Table(s) 1-3, or Table(s) 4-6, or Table(s) 7-9, to identify a compound that specifically binds to the cell-surface receptor or extracellular portion thereof; b) performing a cell-based or biochemical or receptor assay with the compound identified in step a), to identify a compound that modulates one or more non-canonical activities of the ArgRS protein fragment; c) optionally assessing the structure-activity relationship (SAR) of the compound identified in step b), to correlate its structure with modulation of the non-canonical activity, and optionally derivatizing the compound to alter its ability to modulate the non-canonical activity; and d) producing sufficient amounts of the compound identified in step b), or the derivatized compound in step c), for use in humans, thereby manufacturing
- Some embodiments include a cellular composition, comprising an engineered population of cells in which at least one cell comprises a polynucleotide encoding a heterologous full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein, wherein the cells are capable of growing in a serum-free medium.
- the full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein comprises a heterologous purification or epitope tag to facilitate purification of an ArgRS protein fragment.
- the full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein comprises a heterologous proteolysis site to enable production of the ArgRS protein fragment upon cleavage.
- Some embodiments include a method for producing an ArgRS polypeptide as set forth in SEQ ID NO: 29, 151, or 137 in situ within a cell, comprising; i) expressing a heterologous full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein within the cell, wherein the cell comprises a protease capable of cleaving the heterologous full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein to produce the ArgRS polypeptide.
- ArgRS heterologous full length arginyl aminoacyl-tRNA synthetase
- Some embodiments include a method for producing an ArgRS polypeptide as set forth in SEQ ID NO: 29, 151, or 137 comprising contacting an isolated full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein with a protease that is capable of cleaving the full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein and producing an ArgRS polypeptide.
- ArgRS isolated full length arginyl aminoacyl-tRNA synthetase
- Some embodiments include an engineered full length arginyl aminoacyl-tRNA synthetase (ArgRS) protein comprising a heterologous proteolysis site to enable the proteolytic generation of an ArgRS protein fragment as set forth in any of SEQ ID NO: 29, 151, or 137.
- ArgRS arginyl aminoacyl-tRNA synthetase
- Some embodiments include a composition, comprising an isolated full length arginyl aminoacyl-tRNA synthetase protein, wherein the composition has a purity of at least about 95% on a protein basis, less than about 10 EU endotoxin / mg protein, and is substantially serum free.
- the full length arginyl aminoacyl-tRNA synthetase protein is present at a concentration of at least 10 mg / mL, and is at least 90% monodisperse.
- a further embodiment includes a method of treating a disease or disorder mediated by the dysregulation of the expression, activity or spatiotemporal location of a tRNA synthetase via the administration of an ArgRS protein fragment, or nucleic acid encoding the ARRS protein fragment, as set forth in any of SEQ ID NO: 29, 151, or 137.
- the disease is selected cancer, neuropathy, diabetes, and inflammatory disorders.
- the current invention is directed, at least in part, to the discovery of novel ArgRS polypeptides, and methods for their preparation and use, which represent the transformation of native wild type proteins into new forms that exhibit markedly different characteristics compared to the naturally occurring full length Arginyl tRNA synthetase genes.
- Such ArgRS polypeptides were identified based on extensive sequence, and mass spectrometry analysis of expressed Arginyl tRNA synthetase in different tissues, followed by the systematic production and testing of each potential ArgRS polypeptide to identify protein sequences that represent stable and soluble protein domains which exhibit novel biological activities, and favorable therapeutic drug characteristics.
- such Arginyl RNA synthetase derived ArgRS polypeptides comprise polypeptide sequences comprising approximately comprising amino acids 1-71 of Arginyl tRNA synthetase.
- such Arginyl tRNA synthetase derived ArgRS polypeptides comprise polypeptide sequences comprising approximately comprising amino acids 168-530 of Arginyl tRNA synthetase.
- such Arginyl tRNA synthetase derived ArgRS polypeptides comprise polypeptide sequences comprising approximately comprising amino acids 551-660 of Arginyl tRNA synthetase.
- ArgRS polypeptide families represent novel, previously unknown protein products which exhibit inter alia i) novel biological activity, ii) favorable protein stability and aggregation characteristics, and iii) the ability to be expressed and produced at high level in prokaryotic expression systems, which are materially different characteristics not found in the intact wild type protein.
- an element means one element or more than one element.
- An "agonist” refers to a molecule that intensifies or mimics an activity.
- a non-canonical biological activity of an ArgRS or another protein.
- Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition that modulates the activity of an ArgRS either by directly interacting with the ArgRS or its binding partner, or by acting on components of the biological pathway in which the ArgRS participates. Included are partial and full agonists.
- amino acid is intended to mean both naturally occurring and non-naturally occurring amino acids as well as amino acid analogs and mimetics.
- Naturally occurring amino acids include the 20 (L)-amino acids utilized during protein biosynthesis as well as others such as 4-hydroxyproline, hydroxylysine, desmosine, isodesmosine, homocysteine, citrulline and ornithine, for example.
- Non-naturally occurring amino acids include, for example, (D)-amino acids, norleucine, norvaline, p-fluorophenylalanine, ethionine and the like, which are known to a person skilled in the art.
- Amino acid analogs include modified forms of naturally and non-naturally occurring amino acids.
- Such modifications can include, for example, substitution or replacement of chemical groups and moieties on the amino acid or by derivitization of the amino acid.
- Amino acid mimetics include, for example, organic structures which exhibit functionally similar properties such as charge and charge spacing characteristic of the reference amino acid. For example, an organic structure which mimics Arginine (Arg or R) would have a positive charge moiety located in similar molecular space and having the same degree of mobility as the e-amino group of the side chain of the naturally occurring Arg amino acid.
- Mimetics also include constrained structures so as to maintain optimal spacing and charge interactions of the amino acid or of the amino acid functional groups. Those skilled in the art know or can determine what structures constitute functionally equivalent amino acid analogs and amino acid mimetics.
- non-natural amino acids can be utilized to modify (e.g . increase) a selected non-canonical activity of an ArgRS protein fragment, or to alter the in vivo or in vitro half-life of the protein.
- Non-natural amino acids can also be used to facilitate (selective) chemical modifications (e.g ., pegylation) of an ArgRS protein.
- certain non-natural amino acids allow selective attachment of polymers such as PEG to a given protein, and thereby improve their pharmacokinetic properties.
- amino acid analogs and mimetics can be found described in, for example, Roberts and Vellaccio, The Peptides: Analysis, Synthesis, Biology, Eds. Gross and Meinhofer, Vol. 5, p. 341, Academic Press, Inc., New York, N.Y. (1983 ).
- Other examples include peralkylated amino acids, particularly permethylated amino acids. See, for example, Combinatorial Chemistry, Eds. Wilson and Czarnik, Ch. 11, p. 235, John Wiley & Sons Inc., New York, N.Y. (1997 ).
- Yet other examples include amino acids whose amide portion (and, therefore, the amide backbone of the resulting peptide) has been replaced, for example, by a sugar ring, steroid, benzodiazepine or carbo cycle.
- a sugar ring, steroid, benzodiazepine or carbo cycle See, for instance, Burger's Medicinal Chemistry and Drug Discovery, Ed. Manfred E. Wolff, Ch. 15, pp. 619-620, John Wiley & Sons Inc., New York, N.Y. (1995 .
- Methods for synthesizing peptides, polypeptides, peptidomimetics and proteins are well known in the art (see, for example, U.S. Pat. No. 5,420,109 ; M. Bodanzsky, Principles of Peptide Synthesis (1st ed.
- ArgRS polypeptides of the present invention may be composed of naturally occurring and non-naturally occurring amino acids as well as amino acid analogs and mimetics.
- Antagonist refers to a molecule that reduces or attenuates an activity.
- a non-canonical biological activity of an ArgRS or another protein.
- Antagonists may include proteins such as antibodies, nucleic acids, carbohydrates, small molecules, or any other compound or composition that modulates the activity of an ArgRS or its binding partner, either by directly interacting with the ArgRS or its binding partner or by acting on components of the biological pathway in which the ArgRS participates. Included are partial and full antagonists.
- AARS aminoacyl-tRNA synthetase
- aminoacyl-tRNA synthetases catalyze a two-step reaction: first, they activate their respective amino acid by forming an aminoacyl-adenylate, in which the carboxyl of the amino acid is linked in to the alpha-phosphate of ATP by displacing pyrophosphate, and then, when the correct tRNA is bound, the aminoacyl group of the aminoacyl-adenylate is transferred to the 2' or 3' terminal OH of the tRNA.
- Class I aminoacyl-tRNA synthetases typically have two highly conserved sequence motifs. These enzymes aminoacylate at the 2'-OH of an adenosine nucleotide, and are usually monomeric or dimeric. Class II aminoacyl-tRNA synthetases typically have three highly conserved sequence motifs. These enzymes aminoacylate at the 3'-OH of the same adenosine, and are usually dimeric or tetrameric. The active sites of class II enzymes are mainly made up of a seven-stranded antiparallel ⁇ -sheet flanked by ⁇ -helices. Although phenylalanine-tRNA synthetase is class II, it aminoacylates at the 2'-OH.
- AARS polypeptides include sources of mitochondrial and cytoplasmic forms of tyrosyl-tRNA synthetase (TyrRS), a tryptophanyl-tRNA synthetase (TrpRS), a glutaminyl-tRNA synthetase (GlnRS), a glycyl-tRNA synthetase (GlyRS), a histidyl-tRNA synthetase (HisRS), a seryl-tRNA synthetase (SerRS), a phenylalanyl-tRNA synthetase (PheRS), an alanyl-tRNA synthetase (AlaRS), an asparaginyl-tRNA synthetase (AsnRS), an aspartyl-tRNA synthetase (AspRS), a cysteinyl-tRNA synthetase (CysRS), a glutamy
- coding sequence is meant any nucleic acid sequence that contributes to the code for the polypeptide product of a gene.
- non-coding sequence refers to any nucleic acid sequence that does not contribute to the code for the polypeptide product of a gene.
- endotoxin free or “substantially endotoxin free” relates generally to compositions, solvents, and/or vessels that contain at most trace amounts (e.g ., amounts having no clinically adverse physiological effects to a subject) of endotoxin, and preferably undetectable amounts of endotoxin.
- Endotoxins are toxins associated with certain bacteria, typically gram-negative bacteria, although endotoxins may be found in gram-positive bacteria, such as Listeria monocytogenes.
- the most prevalent endotoxins are lipopolysaccharides (LPS) or lipo-oligo-saccharides (LOS) found in the outer membrane of various Gram-negative bacteria, and which represent a central pathogenic feature in the ability of these bacteria to cause disease.
- LPS lipopolysaccharides
- LOS lipo-oligo-saccharides
- ArgRS polypeptides it is often desirable to remove most or all traces of endotoxin from drug products and/or drug containers, because even small amounts may cause adverse effects in humans.
- a depyrogenation oven may be used for this purpose, as temperatures in excess of 300°C are typically required to break down most endotoxins.
- a glass temperature of 250°C and a holding time of 30 minutes is often sufficient to achieve a 3 log reduction in endotoxin levels.
- Other methods of removing endotoxins are contemplated, including, for example, chromatography and filtration methods, as described herein and known in the art.
- compositions comprising ArgRS polypeptides, represent new formulations which exhibit novel and new biological and therapeutic characteristics not found in ArgRS polypeptide compositions contaminated with serum or endotoxin which have the potential to bind to and alter the novel biological properties of the ArgRS polypeptides.
- Endotoxins can be detected using routine techniques known in the art.
- the Limulus Ameobocyte Lysate assay which utilizes blood from the horseshoe crab, is a very sensitive assay for detecting presence of endotoxin, and reagents, kits and instrumentation for the detection of endotoxin based on this assay are commercially available, for example from the Lonza Group.
- very low levels of LPS can cause detectable coagulation of the limulus lysate due a powerful enzymatic cascade that amplifies this reaction.
- Endotoxins can also be quantitated by enzyme-linked immunosorbent assay (ELISA).
- ELISA enzyme-linked immunosorbent assay
- endotoxin levels may be less than about 0.001, 0.005, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.08, 0.09, 0.1, 0.5, 1.0, 1.5, 2, 2.5, 3, 4, 5, 6, 7, 8, 9, or 10 EU /mg of protein.
- 1 ng lipopolysaccharide (LPS) corresponds to about 1-10 EU.
- the "purity" of any given agent (e.g ., ArgRS protein fragment) in a composition may be specifically defined.
- certain compositions may comprise an agent that is at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% pure, including all decimals in between, as measured, for example and by no means limiting, by high pressure liquid chromatography (HPLC), a well-known form of column chromatography used frequently in biochemistry and analytical chemistry to separate, identify, and quantify compounds.
- HPLC high pressure liquid chromatography
- the terms “function” and “functional” and the like refer to a biological, enzymatic, or therapeutic function.
- gene is meant a unit of inheritance that may occupy a specific locus on a chromosome and consists of transcriptional and/or translational regulatory sequences and/or a coding region and/or non-translated sequences (i.e., introns, 5' and 3' untranslated sequences).
- Homology refers to the percentage number of amino acids that are identical or constitute conservative substitutions. Homology may be determined using sequence comparison programs such as GAP ( Deveraux et al., 1984, Nucleic Acids Research, 12, 387-395 ). In this way sequences of a similar or substantially different length to those cited herein could be compared by insertion of gaps into the alignment, such gaps being determined, for example, by the comparison algorithm used by GAP.
- host cell includes an individual cell or cell culture that can be or has been a recipient of any recombinant vector(s), isolated polynucleotide, or polypeptide of the invention.
- Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation and/or change.
- a host cell includes cells transfected or infected in vivo or in vitro with a recombinant vector or a polynucleotide of the invention.
- a host cell which comprises a recombinant vector of the invention is a recombinant host cell.
- isolated is meant material that is substantially or essentially free from components that normally accompany it in its native state.
- an "isolated polynucleotide,” as used herein, includes a polynucleotide that has been purified from the sequences that flank it in its naturally-occurring state, e.g., a DNA fragment which has been removed from the sequences that are normally adjacent to the fragment.
- an "isolated peptide” or an “isolated polypeptide” and the like, as used herein, includes the in vitro isolation and/or purification of a peptide or polypeptide molecule from its natural cellular environment, and from association with other components of the cell; i.e., it is not significantly associated with in vivo substances.
- mRNA or sometimes refer by "mRNA transcripts" as used herein, include, but not limited to pre-mRNA transcript(s), transcript processing intermediates, mature mRNA(s) ready for translation and transcripts of the gene or genes, or nucleic acids derived from the mRNA transcript(s). Transcript processing may include splicing, editing and degradation.
- a nucleic acid derived from an mRNA transcript refers to a nucleic acid for whose synthesis the mRNA transcript or a subsequence thereof has ultimately served as a template.
- mRNA derived samples include, but are not limited to, mRNA transcripts of the gene or genes, cDNA reverse transcribed from the mRNA, cRNA transcribed from the cDNA, DNA amplified from the genes, RNA transcribed from amplified DNA, and the like.
- Non-canonical activity refers generally to either i) a new activity possessed by an ArgRS polypeptide of the invention that is not possessed to any significant degree by the intact native full length parental protein, or ii) an activity that was possessed by the by the intact native full length parental protein, where the ArgRS polypeptide either exhibits a significantly higher (i.e. at least 20% greater) specific activity compared to the intact native full length parental protein, or exhibits the activity in a new context; for example by isolating the activity from other activities possessed by the intact native full length parental protein.
- non-limiting examples of non-canonical activities include extracellular signaling, RNA-binding, amino acid-binding, modulation of cell proliferation, modulation of cell migration, modulation of cell differentiation (e.g ., hematopoiesis, neurogenesis, myogenesis, osteogenesis, and adipogenesis), modulation of gene transcription, modulation of apoptosis or other forms of cell death, modulation of cell signaling, modulation of cellular uptake, or secretion, modulation of angiogenesis, modulation of cell binding, modulation of cellular metabolism, modulation of cytokine production or activity, modulation of cytokine receptor activity, modulation of inflammation, and the like.
- EC 50 half maximal effective concentration refers to the concentration of an ArgRS protein fragment, antibody or other agent described herein at which it induces a response halfway between the baseline and maximum after some specified exposure time; the EC 50 of a graded dose response curve therefore represents the concentration of a compound at which 50% of its maximal effect is observed.
- the EC 50 of an agent provided herein is indicated in relation to a “non-canonical” activity, as noted above.
- EC 50 also represents the plasma concentration required for obtaining 50% of a maximum effect in vivo.
- the “EC 90” refers to the concentration of an agent or composition at which 90% of its maximal effect is observed.
- the "EC 90 " can be calculated from the "EC 50 " and the Hill slope, or it can be determined from the data directly, using routine knowledge in the art.
- the EC 50 of an ArgRS protein fragment, antibody, or other agent is less than about 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, 60, 70, 80, 90, or 100 nM.
- biotherapeutic composition will have an EC 50 value of about 1nM or less.
- modulating includes “increasing” or “stimulating,” as well as “decreasing” or “reducing,” typically in a statistically significant or a physiologically significant amount as compared to a control.
- a “modulator” may be an agonist, an antagonist, or any mixture thereof depending upon the conditions used.
- An “increased” or “enhanced” amount is typically a “statistically significant” amount, and may include an increase that is 1.1, 1.2, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30 or more times ( e.g., 500, 1000 times) (including all integers and decimal points in between and above 1, e.g., 1.5, 1.6, 1.7.
- a "decreased" or reduced amount is typically a "statistically significant” amount, and may include a 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18% , 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% decrease in the amount produced by no composition (the absence of an agent or compound) or a control composition, including all integers in between.
- a control in comparing canonical and non-canonical activities could include the ArgRS protein fragment of interest compared to its corresponding full-length ArgRS, or a fragment ArgRS having comparable canonical activity to its corresponding full-length ArgRS.
- Other examples of "statistically significant" amounts are described herein.
- an ArgRS sequence of the present invention may be "derived” from the sequence information of an ArgRS proteolytic fragment or ArgRS splice variant, or a portion thereof, whether naturally-occurring or artificially generated, and may thus comprise, consist essentially of, or consist of that sequence
- polypeptide and protein are used interchangeably herein to refer to a polymer of amino acid residues and to variants and synthetic and naturally occurring analogues of the same.
- amino acid polymers in which one or more amino acid residues are synthetic non-naturally occurring amino acids, such as a chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally-occurring amino acid polymers and naturally occurring chemical derivatives thereof.
- derivatives include, for example, post-translational modifications and degradation products including pyroglutamyl, iso-aspartyl, proteolytic, phosphorylated, glycosylated, oxidatized, isomerized, and deaminated variants of the ArgRS reference fragment.
- sequence identity or, for example, comprising a “sequence 50% identical to,” as used herein, refer to the extent that sequences are identical on a nucleotide-by-nucleotide basis or an amino acid-by-amino acid basis over a window of comparison.
- a "percentage of sequence identity” may be calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, I) or the identical amino acid residue (e.g ., Ala, Pro, Ser, Thr, Gly, Val, Leu, Ile, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gln, Cys and Met) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison ( i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
- the identical nucleic acid base e.g., A, T, C, G, I
- the identical amino acid residue e.g ., Ala, Pro, Ser, Thr, Gly, Val, Leu, Ile, Phe, Tyr, Trp
- references to describe sequence relationships between two or more polynucleotides or polypeptides include “reference sequence,” “comparison window,” “sequence identity,” “percentage of sequence identity” and “substantial identity.”
- a “reference sequence” is at least 12 but frequently 15 to 18 and often at least 25 monomer units, inclusive of nucleotides and amino acid residues, in length.
- two polynucleotides may each comprise (1) a sequence ( i.e., only a portion of the complete polynucleotide sequence) that is similar between the two polynucleotides, and (2) a sequence that is divergent between the two polynucleotides
- sequence comparisons between two (or more) polynucleotides are typically performed by comparing sequences of the two polynucleotides over a "comparison window" to identify and compare local regions of sequence similarity.
- a “comparison window” refers to a conceptual segment of at least 6 contiguous positions, usually about 50 to about 100, more usually about 100 to about 150 in which a sequence is compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
- the comparison window may comprise additions or deletions ( i.e., gaps) of about 20% or less as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
- Optimal alignment of sequences for aligning a comparison window may be conducted by computerized implementations of algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Drive Madison, WI, USA) or by inspection and the best alignment (i.e., resulting in the highest percentage homology over the comparison window) generated by any of the various methods selected.
- GAP Garnier et al.
- BESTFIT Pearson FASTA
- FASTA Altschul et al.
- TFASTA Pearson's Alignment of Altschul et al.
- a detailed discussion of sequence analysis can be found in Unit 19.3 of Ausubel et al., "Current Protocols in Molecular Biology," John Wiley & Sons Inc, 1994-1998, Chapter 15 .
- sequence similarity or sequence identity between sequences are performed as follows. To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g ., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
- the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, 60%, and even more preferably at least 70%, 80%, 90%, 100% of the length of the reference sequence.
- amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
- the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
- the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
- the percent identity between two amino acid sequences is determined using the Needleman and Wunsch, (1970, J. Mol. Biol. 48: 444-453 ) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
- the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
- a particularly preferred set of parameters are a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frame shift gap penalty of 5.
- the percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of E. Meyers and W. Miller (1989, Cabios, 4: 11-17 ) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
- nucleic acid and protein sequences described herein can be used as a "query sequence" to perform a search against public databases to, for example, identify other family members or related sequences.
- Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al., (1990, J. Mol. Biol, 215: 403-10 ).
- Gapped BLAST can be utilized as described in Altschul et al., (1997, Nucleic Acids Res, 25: 3389-3402 ).
- the default parameters of the respective programs e.g ., XBLAST and NBLAST
- the default parameters of the respective programs e.g ., XBLAST and NBLAST
- solubility refers to the property of an agent provided herein to dissolve in a liquid solvent and form a homogeneous solution. Solubility is typically expressed as a concentration, either by mass of solute per unit volume of solvent (g of solute per kg of solvent, g per dL (100 mL), mg/ml, etc.), molarity, molality, mole fraction or other similar descriptions of concentration.
- the maximum equilibrium amount of solute that can dissolve per amount of solvent is the solubility of that solute in that solvent under the specified conditions, including temperature, pressure, pH, and the nature of the solvent.
- solubility is measured at physiological pH. In certain embodiments, solubility is measured in water or a physiological buffer such as PBS.
- solubility is measured in a biological fluid (solvent) such as blood or serum.
- the temperature can be about room temperature (e.g ., about 20, 21, 22, 23, 24, 25°C) or about body temperature (37°C).
- an agent such as an ArgRS protein fragment has a solubility of at least about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, or 30 mg/ml at room temperature or at 37°C.
- a "splice junction" as used herein includes the region in a mature mRNA transcript or the encoded polypeptide where the 3' end of a first exon joins with the 5' end of a second exon.
- the size of the region may vary, and may include 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more (including all integers in between) nucleotide or amino acid residues on either side of the exact residues where the 3' end of one exon joins with the 5' end of another exon.
- an “exon” refers to a nucleic acid sequence that is represented in the mature form of an RNA molecule after either portions of a precursor RNA (introns) have been removed by cis-splicing or two or more precursor RNA molecules have been ligated by trans-splicing.
- the mature RNA molecule can be a messenger RNA or a functional form of a non-coding RNA such as rRNA or tRNA.
- an exon can refer to the sequence in the DNA or its RNA transcript.
- An "intron” refers to a non-coding nucleic acid region within a gene, which is not translated into a protein. Non-coding intronic sections are transcribed to precursor mRNA (pre-mRNA) and some other RNAs (such as long noncoding RNAs), and subsequently removed by splicing during the processing to mature RNA.
- a “splice variant” refers to a mature mRNA and its encoded protein that are produced by alternative splicing, a process by which the exons of the RNA (a primary gene transcript or pre-mRNA) are reconnected in multiple ways during RNA splicing. The resulting different mRNAs may be translated into different protein isoforms, allowing a single gene to code for multiple proteins.
- a "subject,” as used herein, includes any animal that exhibits a symptom, or is at risk for exhibiting a symptom, which can be treated or diagnosed with an ArgRS polynucleotide or polypeptide of the invention. Also included are subjects for which it is desirable to profile levels of ArgRS polypeptides and/or polynucleotides of the invention, for diagnostic or other purposes. Suitable subjects (patients) include laboratory animals (such as mouse, rat, rabbit, or guinea pig), farm animals, and domestic animals or pets (such as a cat or dog). Non-human primates and, preferably, human patients, are included.
- Treatment includes any desirable effect on the symptoms or pathology of a disease or condition that can be effected by the non-canonical activities of an ArgRS polynucleotide or polypeptide, as described herein, and may include even minimal changes or improvements in one or more measurable markers of the disease or condition being treated. Also included are treatments that relate to non-ArgRS therapies, in which an ArgRS sequence described herein provides a clinical marker of treatment. "Treatment” or “treating” does not necessarily indicate complete eradication or cure of the disease or condition, or associated symptoms thereof. The subject receiving this treatment is any subject in need thereof. Exemplary markers of clinical improvement will be apparent to persons skilled in the art.
- ArgRS fragments possess biological activities important for biotherapeutic, discovery and diagnostic applications.
- Embodiments of the present invention therefore include full length proteins, mature protein isoforms and protein fragments of arginyl aminoacyl-tRNA synthetase (ArgRS), in addition to biologically active variants and fragments thereof.
- the proteins and fragments may arise through endogenous proteolysis, in vitro proteolysis, splice variation, or in silico prediction, among other mechanisms.
- the ArgRS protein fragments described herein, and variants thereof, may possess at least one "non-canonical" biological activity.
- the ArgRS protein fragment(s) of the present invention are also referred to herein as "ArgRS polypeptides" or "ArgRS reference polypeptides.”
- the ArgRS polypeptides provided herein comprise or consist essentially of all or a portion of the ArgRS polypeptide "reference sequence(s)" as set forth in SEQ ID NO: 29, 151, or 137 below, which represent the amino acid sequence(s) of various fragments of Arginyl tRNA synthetases.
- Mouse and human AARS protein sequences are highly related, typically differing by no more than a few amino acids within an entire sequence, a particular domain, or a particular protein fragment.
- N-terminal ArgRS Polypeptides (Tables 1, 2 & 3) Table 1A ArgRS polypeptides identified by MS Name Type / species / Residues Amino acid and Nucleic Acid Sequences SEQ.ID . NO. ArgRS N1 Protein / Human / 1-398 SEQ.ID. NO.12 ArgRS N1 DNA / Human / SEQ.ID. NO.13 Table 1B ArgRS N1 Mass spec peptides detected and inferred linking peptides Type / species Sequence SEQ.ID . NO.
- Protein / mouse LQEVFGCAIR SEQ.ID. NO.45 Protein / mouse AAYPDLENPPLIVTPSQQPK SEQ.ID. NO.46 Protein / mouse FGDYQCN SEQ.ID. NO.47 Protein / mouse SEQ.ID. NO.48 Protein / mouse VIVDFSSPNIAK SEQ.ID. NO.49 Protein / mouse EMHVGHLRSTIIGESMSR SEQ.ID. NO.50 Protein / mouse LFEFAGYDVLR SEQ.ID. NO.51 Protein / mouse LNHVGDWGTQFGMLIAHLQDK SEQ.ID. NO.52 Protein / mouse FPDYLTVSPPIGDLQAFYK SEQ.ID.
- Protein fragments or the amino acid sequence of protein fragments, such as proteolytic fragments or splice variant fragments, can be characterized, identified, or derived according to a variety of techniques. For instance, splice variants can be identified by techniques such as deep sequencing (see, e.g., Xing et al., RNA. 14:1470-1479, 2008 ; and Zhang et al., Genome Research,. 17:503-509, 2007 ). As a further example, protein fragments such as proteolytic fragments can be identified in vitro, such as by incubating full-length or other ArgRS polypeptides with selected proteases, or they can be identified endogenously ( e.g., in vivo ).
- protein fragments such as endogenous proteolytic fragments can be generated or identified, for instance, by recombinantly expressing full-length or other ArgRS polypeptides in a selected microorganism or eukaryotic cell that has been either modified to contain one or more selected proteases, or that naturally contains one or more proteases that are capable of acting on a selected ArgRS polypeptide, and isolating and characterizing the endogenously produced protein fragments therefrom.
- protein fragments such as endogenous (e.g ., naturally-occurring) proteolytic fragments can be generated or identified, for instance, from various cellular fractions (e.g ., cytosolic, membrane, nuclear) and/or growth medium of various cell-types, including, for example, immune cells such as monocytes, dendritic cells, macrophages ( e.g ., RAW 264.7 macrophages), neutrophils, eosinophils, basophils, and lymphocytes, such as B-cells and T-cells (e.g., CD4+ helper and CD8+ killer cells), including primary T-cells and T-cell lines such as Jurkat T-cells, as well as natural killer (NK) cells.
- immune cells such as monocytes, dendritic cells, macrophages (e.g ., RAW 264.7 macrophages), neutrophils, eosinophils, basophils, and lymphocytes, such as B-cells and T
- protein fragments such as endogenous proteolytic fragments, however generated, can be identified by techniques such as mass-spectrometry, or equivalent techniques. Once an in vitro or endogenously identified protein fragment has been generated or identified, it can be mapped or sequenced, and, for example, cloned into an expression vector for recombinant production, or produced synthetically.
- proteases can be used to produce, identify, derive, or characterize the sequence of ArgRS protein fragments such as proteolytic fragments.
- proteases are usually classified according to three major criteria: (i) the reaction catalyzed, (ii) the chemical nature of the catalytic site, and (iii) the evolutionary relationship, as revealed by the structure.
- General examples of proteases or proteinases, as classified by mechanism of catalysis, include aspartic proteases, serine proteases, cysteine proteases, and metalloproteases.
- aspartic proteases belong to the pepsin family. This family includes digestive enzymes, such as pepsin and chymosin, as well as lysosomal cathepsins D and processing enzymes such as renin, and certain fungal proteases (e.g ., penicillopepsin, rhizopuspepsin, endothiapepsin).
- a second family of aspartic proteases includes viral proteinases such as the protease from the AIDS virus (HIV), also called retropepsin.
- Serine proteases include two distinct families. First, the chymotrypsin family, which includes the mammalian enzymes such as chymotrypsin, trypsin, elastase, and kallikrein, and second, the substilisin family, which includes the bacterial enzymes such as subtilisin.
- the general 3D structure between these two families is different, but they have the same active site geometry, and catalysis proceeds via the same mechanism.
- the serine proteases exhibit different substrate specificities, differences which relate mainly to amino acid substitutions in the various enzyme subsites (substrate residue interacting sites). Some serine proteases have an extended interaction site with the substrate whereas others have a specificity that is restricted to the P1 substrate residue.
- the cysteine protease family includes the plant proteases such as papain, actinidin, and bromelain, several mammalian lysosomal cathepsins, the cytosolic calpains (calcium-activated), as well as several parasitic proteases ( e.g., Trypanosoma, Schistosoma ).
- Papain is the archetype and the best studied member of the family. Recent elucidation of the X-ray structure of the Interleukin-1-beta Converting Enzyme has revealed a novel type of fold for cysteine proteinases.
- the metalloproteases are one of the older classes of proteases, found in bacteria, fungi, and higher organisms. They differ widely in their sequences and their 3D structures, but the great majority of enzymes contain a zinc atom that is catalytically active. In some cases, zinc may be replaced by another metal such as cobalt or nickel without loss of proteolytic activity.
- Bacterial thermolysin has been well characterized and its crystallographic structure indicates that zinc is bound by two histidines and one glutamic acid.
- Many metalloproteases contain the sequence motif HEXXH, which provides two histidine ligands for the zinc. The third ligand is either a glutamic acid (thermolysin, neprilysin, alanyl aminopeptidase) or a histidine (astacin, serralysin).
- Illustrative proteases include, for example, achromopeptidase, aminopeptidase, ancrod, angiotensin converting enzyme, bromelain, calpain, calpain I, calpain II, carboxypeptidase A, carboxypeptidase B, carboxypeptidase G, carboxypeptidase P, carboxypeptidase W, carboxypeptidase Y, caspase 1, caspase 2, caspase 3, caspase 4, caspase 5, caspase 6, caspase 7, caspase 8, caspase 9, caspase 10, caspase 11, caspase 12, caspase 13, cathepsin B, cathepsin C, cathepsin D, cathepsin E, cathepsin G, cathepsin H, cathepsin L, chymopapain , chymase, chymotrypsin, clostripain, collagenase, complement C1r, complement C1
- protease from Bacillus polymyxa protease from Bacillus sp, protease from Rhizopus sp., protease S, proteasomes, proteinase from Aspergillus oryzae, proteinase 3, proteinase A, proteinase K, protein C, pyroglutamate aminopeptidase, rennin, rennin, streptokinase, subtilisin, thermolysin, thrombin, tissue plasminogen activator, trypsin, tryptase and urokinase.
- ArgRS polypeptides comprising, consisting essentially of, or consisting of amino acid sequences that have been derived from endogenous, naturally-occurring ArgRS polypeptide fragments, and pharmaceutical compositions comprising said fragments, and methods of use thereof.
- ArgRS proteolytic fragments are generated or identified by incubating an ArgRS polypeptide, such as a full-length ArgRS polypeptide, with one or more isolated human proteases, mainly those proteases that are endogenous or natural to humans, such as elastase and others described herein and known in the art.
- ArgRS polypeptides comprising, consisting essentially of, or consisting of amino acid sequences that have been derived from endogenous, naturally-occurring ArgRS splice variants, and pharmaceutical compositions comprising said fragments, and methods of use thereof.
- ArgRS protein fragment can be isolated from samples that have been exposed to proteases, whether in vivo or in vitro.
- ArgRS protein fragments can be identified by techniques such as mass-spectrometry, or equivalent techniques.
- the proteomes from various cell types, tissues, or body fluids from a variety of physiological states e.g ., hypoxia, diet, age, disease
- the proteomes from a variety of physiological states e.g ., hypoxia, diet, age, disease
- fractions thereof may be separated by 1D SDS-PAGE and the gel lanes cut into bands at fixed intervals; after which the bands may be optionally digested with an appropriate protease, such as trypsin, to release the peptides, which may then be analyzed by 1D reverse phase LC-MS/MS.
- the resulting proteomic data may be integrated into so-called peptographs, which plot, in the left panel, sequence coverage for a given protein in the horizontal dimension (N to C terminus, left to right) versus SDS-PAGE migration in the vertical dimension (high to low molecular weight, top to bottom).
- the specific peptide fragments can then be sequenced or mapped.
- the ArgRS reference fragment may be characterized by its unique molecular weight, as compared, for example, to the molecular weight of the corresponding full-length ArgRS.
- embodiments of the present invention include the ArgRS polypeptides set forth in SEQ ID NO: 29, 151, or 137. Also included are “variants” of the ArgRS reference polypeptides.
- the recitation polypeptide "variant” refers to polypeptides that are distinguished from a reference ArgRS polypeptide by the addition, deletion, and/or substitution of at least one amino acid residue, and which typically retain (e.g., mimic) or modulate (e.g., antagonize) one or more non-canonical activities of a reference ArgRS polypeptide.
- human Arginyl tRNA synthetases include several hundred highly related polymorphic forms, and these are known in the art to be at least partially functionally interchangeable. It would thus be a routine matter to select a naturally occurring variant of Arginyl tRNA synthetase, including, for example the single nucleotide polymorphic forms listed in Table A to create an ArgRS polypeptide containing one or more amino acid changes based on the sequence of any of the homologues, orthologs, and naturally-occurring isoforms of human as well as other species of Arginyl tRNA synthetase.
- a polypeptide variant is distinguished from a reference polypeptide by one or more substitutions, which may be conservative or non-conservative, as described herein and known in the art.
- the polypeptide variant comprises conservative substitutions and, in this regard, it is well understood in the art that some amino acids may be changed to others with broadly similar properties without changing the nature of the activity of the polypeptide.
- a variant polypeptide includes an amino acid sequence having at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or more sequence identity or similarity to a corresponding sequence of an ArgRS reference polypeptide, as described herein, and substantially retains the non-canonical activity of that reference polypeptide.
- sequences differing from the reference ArgRS sequences by the addition, deletion, or substitution of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60 ,70, 80, 90, 100, 110, 120, 130, 140, 150 or more amino acids but which retain the properties of the reference ArgRS polypeptide.
- the amino acid additions or deletions occur at the C-terminal end and/or the N-terminal end of the ArgRS reference polypeptide.
- the amino acid additions include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50 or more wild-type residues (i.e ., from the corresponding full-length ArgRS polypeptide) that are proximal to the C-terminal end and/or the N-terminal end of the ArgRS reference polypeptide.
- variant polypeptides differ from the corresponding ArgRS reference sequences by at least 1% but less than 20%, 15%, 10% or 5% of the residues. (If this comparison requires alignment, the sequences should be aligned for maximum similarity. "Looped" out sequences from deletions or insertions, or mismatches, are considered differences.) The differences are, suitably, differences or changes at a non-essential residue or a conservative substitution.
- the molecular weight of a variant ArgRS polypeptide differs from that of the ArgRS reference polypeptide by about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, or more.
- biologically active fragments of the ArgRS reference polypeptides, i.e., biologically active fragments of the ArgRS protein fragments.
- Representative biologically active fragments generally participate in an interaction, e.g ., an intramolecular or an inter-molecular interaction.
- An inter-molecular interaction can be a specific binding interaction or an enzymatic interaction.
- An inter-molecular interaction can be between an ArgRS polypeptide and a cellular binding partner, such as a cellular receptor or other host molecule that participates in the non-canonical activity of the ArgRS polypeptide.
- ArgRS proteins, variants, and biologically active fragments thereof bind to one or more cellular binding partners with an affinity of at least about 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, or 50 nM.
- the binding affinity of an ArgRS protein fragment for a selected cellular binding partner, particularly a binding partner that participates in a non-canonical activity, is typically stronger than that of the ArgRS protein fragment's corresponding full-length ArgRS polypeptide, by at least about 1.5x, 2x, 2.5x, 3x, 3.5x, 4x, 4.5x, 5x, 6x, 7x, 8x, 9x, 10x, 15x, 20x, 25x, 30x, 40x, 50x, 60x, 70x, 80x, 90x, 100x, 200x, 300x, 400x, 500x, 600x, 700x, 800x, 900x, 1000x or more (including all integers in between).
- the binding affinity of an ArgRS protein fragment for a binding partner that participates in at least one canonical activity of an ArgRS is typically weaker than that of the ArgRS protein fragment's corresponding full-length ArgRS polypeptide, by at least about 1.5x, 2x, 2.5x, 3x, 3.5x, 4x, 4.5x, 5x, 6x, 7x, 8x, 9x, 10x, 15x, 20x, 25x, 30x, 40x, 50x, 60x, 70x, 80x, 90x, 100x, 200x, 300x, 400x, 500x, 600x, 700x, 800x, 900x, 1000x or more.
- biologically active fragments comprise a domain or motif with at least one activity of an ArgRS reference polypeptide and may include one or more (and in some cases all) of the various active domains, and include fragments having a non-canonical activity.
- biologically active fragments of an ArgRS polypeptide have a biological activity that is unique to the particular, truncated fragment, such that the full-length ArgRS polypeptide may not have that activity.
- the biological activity may be revealed by separating the biologically active ArgRS polypeptide fragment from the other full-length ArgRS polypeptide sequences, or by altering certain residues of the full-length ArgRS wild-type polypeptide sequence to unmask the biologically active domains.
- a biologically active fragment of an ArgRS reference polypeptide can be a polypeptide fragment which is, for example, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400, 450, 500, 550, 600, 650, 700, 750 or more contiguous or non-contiguous amino acids, including all integers ( e.g., 101, 102, 103) and ranges ( e.g., 50-100, 50-150, 50-200) in between, of the amino acid sequences set forth in any one of the ArgRS reference polypeptides described herein, but typically exclude the full-length ArgRS.
- a biologically active fragment comprises a non-canonical activity-related sequence, domain, or motif.
- the C-terminal or N-terminal region of any ArgRS reference polypeptide may be truncated by about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, or 700 or more amino acids, or by about 10-50, 20-50, 50-100, 100-150, 150-200, 200-250, 250-300, 300-350, 350-400, 400-450, 450-500, 500-550, 550-600, 600-650, 650-700 or more amino acids, including all integers and ranges in between (e.g., 101, 102, 103, 104, 105), so long as the truncated ArgRS poly
- the biologically-active fragment has no less than about 1%, about 5 %, about 10%, about 25%, or about 50% of an activity of the biologically-active (i.e ., non-canonical activity) ArgRS reference polypeptide from which it is derived. Exemplary methods for measuring such non-canonical activities are described in the Examples.
- an ArgRS polypeptide may be altered in various ways including amino acid substitutions, deletions, truncations, and insertions. Methods for such manipulations are generally known in the art.
- amino acid sequence variants of an ArgRS reference polypeptide can be prepared by mutations in the DNA. Methods for mutagenesis and nucleotide sequence alterations are well known in the art. See, for example, Kunkel (1985, Pro. Natl. Acad. Sci. USA. 82: 488-492 ), Kunkel et al., (1987, Methods in Enzymol, 154: 367-382 ), U.S. Pat. No. 4,873,192 , Watson, J. D.
- Biologically active truncated and/or variant ArgRS polypeptides may contain conservative amino acid substitutions at various locations along their sequence, as compared to a reference ArgRS amino acid residue.
- naturally occurring variants of ArgRS proteins have been sequenced, and are known in the art to be at least partially functionally interchangeable. It would thus be a routine matter to select an amino acid position to introduce a conservative, or non-conservative mutation into an ArgRS polypeptide based on naturally occurring sequence variation among the known ArgRS protein homologues, orthologs, and naturally-occurring isoforms of human as well as other species of an ArgRS protein.
- a “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, which can be generally sub-classified as follows:
- Acidic The residue has a negative charge due to loss of H ion at physiological pH and the residue is attracted by aqueous solution so as to seek the surface positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH.
- Amino acids having an acidic side chain include glutamic acid and aspartic acid.
- the residue has a positive charge due to association with H ion at physiological pH or within one or two pH units thereof (e.g., histidine) and the residue is attracted by aqueous solution so as to seek the surface positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium at physiological pH.
- Amino acids having a basic side chain include arginine, lysine and histidine.
- the residues are charged at physiological pH and, therefore, include amino acids having acidic or basic side chains (i.e ., glutamic acid, aspartic acid, arginine, lysine and histidine).
- amino acids having acidic or basic side chains i.e ., glutamic acid, aspartic acid, arginine, lysine and histidine.
- Hydrophobic The residues are not charged at physiological pH and the residue is repelled by aqueous solution so as to seek the inner positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium.
- Amino acids having a hydrophobic side chain include tyrosine, valine, isoleucine, leucine, methionine, phenylalanine and tryptophan.
- Neutral/polar The residues are not charged at physiological pH, but the residue is not sufficiently repelled by aqueous solutions so that it would seek inner positions in the conformation of a peptide in which it is contained when the peptide is in aqueous medium.
- Amino acids having a neutral/polar side chain include asparagine, glutamine, cysteine, histidine, serine and threonine.
- proline This description also characterizes certain amino acids as “small” since their side chains are not sufficiently large, even if polar groups are lacking, to confer hydrophobicity.
- "small” amino acids are those with four carbons or less when at least one polar group is on the side chain and three carbons or less when not.
- Amino acids having a small side chain include glycine, serine, alanine and threonine.
- the gene-encoded secondary amino acid proline is a special case due to its known effects on the secondary conformation of peptide chains.
- the structure of proline differs from all the other naturally-occurring amino acids in that its side chain is bonded to the nitrogen of the ⁇ -amino group, as well as the ⁇ -carbon.
- the degree of attraction or repulsion required for classification as polar or nonpolar is arbitrary and, therefore, amino acids specifically contemplated by the invention have been classified as one or the other. Most amino acids not specifically named can be classified on the basis of known behavior.
- Amino acid residues can be further sub-classified as cyclic or non-cyclic, and aromatic or non-aromatic, self-explanatory classifications with respect to the side-chain substituent groups of the residues, and as small or large.
- the residue is considered small if it contains a total of four carbon atoms or less, inclusive of the carboxyl carbon, provided an additional polar substituent is present; three or less if not.
- Small residues are, of course, always non-aromatic.
- amino acid residues may fall in two or more classes. For the naturally-occurring protein amino acids, sub-classification according to this scheme is presented in Table B.
- Table B Amino acid sub-classification Sub-classes Amino acids Acidic Aspartic acid, Glutamic acid Basic Noncyclic: Arginine, Lysine; Cyclic: Histidine Charged Aspartic acid, Glutamic acid, Arginine, Lysine, Histidine Small Glycine, Serine, Alanine, Threonine, Proline Polar/neutral Asparagine, Histidine, Glutamine, Cysteine, Serine, Threonine Polar/large Asparagine, Glutamine Hydrophobic Tyrosine, Valine, Isoleucine, Leucine, Methionine, Phenylalanine, Tryptophan Aromatic Tryptophan, Tyrosine, Phenylalanine Residues that influence chain orientation Glycine and Proline
- Conservative amino acid substitution also includes groupings based on side chains.
- a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulfur-containing side chains is cysteine and methionine.
- Amino acid substitutions falling within the scope of the invention are, in general, accomplished by selecting substitutions that do not differ significantly in their effect on maintaining (a) the structure of the peptide backbone in the area of the substitution, (b) the charge or hydrophobicity of the molecule at the target site, (c) the bulk of the side chain, or (d) the biological function. After the substitutions are introduced, the variants are screened for biological activity.
- similar amino acids for making conservative substitutions can be grouped into three categories based on the identity of the side chains.
- the first group includes glutamic acid, aspartic acid, arginine, lysine, histidine, which all have charged side chains;
- the second group includes glycine, serine, threonine, cysteine, tyrosine, glutamine, asparagine;
- the third group includes leucine, isoleucine, valine, alanine, proline, phenylalanine, tryptophan, methionine, as described in Zubay, G., Biochemistry , third edition, Wm.C. Brown Publishers (1993).
- a predicted non-essential amino acid residue in a truncated and/or variant ArgRS polypeptide is typically replaced with another amino acid residue from the same side chain family.
- mutations can be introduced randomly along all or part of an ArgRS coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for an activity of the parent polypeptide to identify mutants which retain that activity.
- the encoded peptide can be expressed recombinantly and the activity of the peptide can be determined.
- a "non-essential" amino acid residue is a residue that can be altered from the reference sequence of an embodiment polypeptide without abolishing or substantially altering one or more of its activities.
- the alteration does not substantially abolish one of these activities, for example, the activity is at least 20%, 40%, 60%, 70% or 80% 100%, 500%, 1000% or more of the reference ArgRS sequence.
- An "essential" amino acid residue is a residue that, when altered from the reference sequence of an ArgRS polypeptide, results in abolition of an activity of the parent molecule such that less than 20% of the reference activity is present.
- such essential amino acid residues include those that are conserved in ArgRS polypeptides across different species, including those sequences that are conserved in the active binding site(s) or motif(s) of ArgRS polypeptides from various sources.
- polypeptides and fusion polypeptides are isolated.
- An "isolated" polypeptide or polynucleotide is one that is removed from its original environment.
- a naturally-occurring protein is isolated if it is separated from some or all of the coexisting materials in the natural system.
- polypeptides are at least about 90% pure, more preferably at least about 95% pure and most preferably at least about 99% pure.
- a polynucleotide is considered to be isolated if, for example, it is cloned into a vector that is not a part of the natural environment.
- Dimers may include, for example, homodimers between two identical ArgRS polypeptides, heterodimers between two different ArgRS polypeptides (e.g., a full-length YRS polypeptide and a truncated YRS polypeptide; a truncated YRS polypeptide and a truncated WRS polypeptide), and/or heterodimers between an ArgRS polypeptide and a heterologous polypeptide.
- Certain heterodimers, such as those between an ArgRS polypeptide and a heterologous polypeptide may be bi-functional, as described herein.
- monomers of ArgRS polypeptides including isolated ArgRS polypeptides monomers that do not substantially dimerize with a second ArgRS polypeptide, whether due to one or more substitutions, truncations, deletions, additions, chemical modifications, or a combination of these alterations.
- monomeric ArgRS polypeptides possess biological activities, including non-canonical activities, which are not possessed by dimeric or multimeric ArgRS polypeptide complexes.
- ArgRS polypeptides include modifications that improved the desired characteristics of an ArgRS polypeptide, as described herein.
- Modifications of ArgRS polypeptides of the invention include chemical and/or enzymatic derivatizations at one or more constituent amino acid, including side chain modifications, backbone modifications, and N- and C-terminal modifications including acetylation, hydroxylation, methylation, amidation, and the attachment of carbohydrate or lipid moieties, cofactors, and the like.
- Exemplary modifications also include pegylation of an ArgRS polypeptide (see, e.g., Veronese and Harris, Advanced Drug Delivery Reviews 54: 453-456, 2002 ; and Pasut et al., Expert Opinion. Ther. Patents 14(6) 859-894 2004 ).
- PEG is a well-known polymer having the properties of solubility in water and in many organic solvents, lack of toxicity, and lack of immunogenicity. It is also clear, colorless, odorless, and chemically stable. For these reasons and others, PEG has been selected as the preferred polymer for attachment, but it has been employed solely for purposes of illustration and not limitation.
- Similar products may be obtained with other water-soluble polymers, including without limitation; polyvinyl alcohol, other poly(alkylene oxides) such as poly(propylene glycol) and the like, poly(oxyethylated polyols) such as poly(oxyethylated glycerol) and the like, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl purrolidone, poly-1,3- dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride, and polyaminoacids.
- polyvinyl alcohol other poly(alkylene oxides) such as poly(propylene glycol) and the like
- poly(oxyethylated polyols) such as poly(oxyethylated glycerol) and the like
- carboxymethylcellulose carboxymethylcellulose
- dextran polyvinyl alcohol
- polyvinyl purrolidone poly-1,3- dioxolane
- PEG derivatives are both available and suitable for use in the preparation of PEG-conjugates.
- NOF Corp.'s PEG reagents sold under the trademark SUNBRIGHT® Series provides numerous PEG derivatives, including methoxypolyethylene glycols and activated PEG derivatives such as methoxy-PEG amines, maleimides, N-hydroxysuccinimide esters, and carboxylic acids, for coupling by various methods to the N-terminal, C-terminal or any internal amino acid of the ArgRS polypeptide.
- Nektar Therapeutics' Advanced PEGylation technology also offers diverse PEG-coupling technologies to potentially improve the safety and efficacy of an ArgRS polypeptide based therapeutic.
- chemoselective ligation technology may be utilized to modify ArgRS polypeptides of the invention, such as by attaching polymers in a site-specific and controlled manner.
- Such technology typically relies on the incorporation of chemoselective anchors into the protein backbone by either chemical or recombinant means, and subsequent modification with a polymer carrying a complementary linker.
- the assembly process and the covalent structure of the resulting protein-polymer conjugate may be controlled, enabling the rational optimization of drug properties, such as efficacy and pharmacokinetic properties ( see, e.g., Kochendoerfer, Current Opinion in Chemical Biology 9:555-560, 2005 ).
- fusion proteins of ArgRS polypeptide to other proteins are also included, and these fusion proteins may increase the ArgRS polypeptide's biological activity, secretion, targeting, biological life, the ability to penetrate cellular membranes or the blood brain barrier, or pharmacokinetic properties.
- fusion proteins that improve pharmacokinetic properties include without limitation, fusions to human albumin ( Osborn et al.: Eur. J. Pharmacol. 456(1-3): 149-158, (2002 )), antibody Fc domains, poly Glu or poly Asp sequences, and transferrin.
- fusion with conformationally disordered polypeptide sequences composed of the amino acids Pro, Ala, and Ser ('PASylation') or hydroxyethyl starch (sold under the trademark HESYLATION®) provides a simple way to increase the hydrodynamic volume of the ArgRS polypeptide.
- This additional extension adopts a bulky random structure, which significantly increases the size of the resulting fusion protein. By this means the typically rapid clearance of smaller ArgRS polypeptides via kidney filtration is retarded by several orders of magnitude.
- Ig G fusion proteins has also been shown to enable some fusion protein proteins to penetrate the blood brain barrier ( Fu et al., (2010) Brain Res. 1352:208-13 ).
- membrane translocating sequences refers to naturally occurring and synthetic amino acid sequences that are capable of membrane translocation across a cellular membrane.
- Representative membrane translocating sequences include those based on the naturally occurring membrane translocating sequences derived from the Tat protein, and homeotic transcription protein Antennapedia, as well as synthetic membrane translocating sequences based in whole or part on poly Arginine and Lysine resides.
- Representative membrane translocating sequences include for example those disclosed in the following patents, US5,652,122 ; US 5,670,617 ; US5,674,980 ; US5,747,641 ; US5,804,604 ; US6,316,003 ; US7,585,834 ; US7,312,244 ; US7,279,502 ; US7,229,961 ; US7,169,814 ; US7,453,011 ; US7,235,695 ; US6,982,351 ; US6,605,115 ; US7,306,784 ; US7,306,783 ; US6,589,503 ; US6,348,185 ; US6,881,825 ; US7,431,915 ; WO0074701A2 ; WO2007111993A2 ; WO2007106554A2 ; WO02069930A1 ; WO03049772A2 ; WO03106491A
- a flexible molecular linker (or spacer) optionally may be interposed between, and covalently join, the ArgRS polypeptide and any of the fusion proteins disclosed herein.
- the ArgRS polypeptide can include synthetic, or naturally occurring secretion signal sequences, derived from other well characterized secreted proteins. In some embodiments such proteins, may be processed by proteolytic cleavage to form the ArgRS polypeptide in situ.
- fusions proteins include for example fusions of ArgRS polypeptide to ubiquitin to provide a new N-terminal amino acid, or the use of a secretion signal to mediate high level secretion of the ArgRS polypeptide into the extracellular medium, or N, or C-terminal epitope tags to improve purification or detection.
- ArgRS polypeptides described herein may be prepared by any suitable procedure known to those of skill in the art, such as by recombinant techniques.
- polypeptides of the invention may be produced by direct peptide synthesis using solid-phase techniques (Merrifield, J. Am. Chem. Soc. 85 :2149-2154 (1963)). Protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer). Alternatively, various fragments may be chemically synthesized separately and combined using chemical methods to produce the desired molecule.
- Embodiments of the present invention include polynucleotides that encode one or more newly identified protein fragments of an arginyl aminoacyl-tRNA synthetase (ArgRS), in addition to complements, variants, and fragments thereof.
- an ArgRS polynucleotide encodes all or a portion of the ArgRS polypeptide reference sequence(s) as set forth in SEQ ID NO: 29, 151, or 137, which represent fragments of Arginyl tRNA synthetase.
- ArgRS polynucleotides of the present invention are primers, probes, antisense oligonucleotides, and RNA interference agents that comprise all or a portion of these reference polynucleotides, which are complementary to all or a portion of these reference polynucleotides, or which specifically hybridize to these reference polynucleotides, as described herein.
- polynucleotide or “nucleic acid” as used herein designates mRNA, RNA, cRNA, cDNA or DNA.
- the term typically refers to polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide.
- the term includes single and double stranded forms of DNA.
- DNA and “polynucleotide” and “nucleic acid” refer to a DNA molecule that has been isolated free of total genomic DNA of a particular species.
- an isolated DNA segment encoding a polypeptide refers to a DNA segment that contains one or more coding sequences yet is substantially isolated away from, or purified free from, total genomic DNA of the species from which the DNA segment is obtained. Also included are non-coding polynucleotides (e.g., primers, probes, oligonucleotides), which do not encode an ArgRS polypeptide. Included within the terms "DNA segment” and “polynucleotide” are DNA segments and smaller fragments of such segments, and also recombinant vectors, including, for example, plasmids, cosmids, phagemids, phage, viruses, and the like.
- Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
- the polynucleotides of the present invention regardless of the length of the coding sequence itself, may be combined with other DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably.
- polynucleotide fragment of almost any length may be employed; with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol. Included are polynucleotides of about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 41, 43, 44, 45, 46, 47, 48, 49, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 220, 240, 260, 270, 280, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800
- Embodiments of the present invention also include "variants" of the ArgRS reference polynucleotide sequences.
- Polynucleotide "variants" may contain one or more substitutions, additions, deletions and/or insertions in relation to a reference polynucleotide.
- variants of an ArgRS reference polynucleotide sequence may have at least about 30%, 40% 50%, 55%, 60%, 65%, 70%, generally at least about 75%, 80%, 85%, desirably about 90% to 95% or more, and more suitably about 98% or more sequence identity to that particular nucleotide sequence as determined by sequence alignment programs described elsewhere herein using default parameters.
- variants may differ from a reference sequence by about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 41, 43, 44, 45, 46, 47, 48, 49, 50, 60, 70, 80, 90, 100 (including all integers in between) or more bases.
- the polynucleotide variant encodes an ArgRS polypeptide having a non-canonical activity
- the desired activity of the encoded ArgRS polypeptide is not substantially diminished relative to the unmodified polypeptide.
- the effect on the activity of the encoded polypeptide may generally be assessed as described herein.
- Certain embodiments include polynucleotides that hybridize to a reference ArgRS polynucleotide sequence, or to their complements, under stringency conditions described below.
- the term “hybridizes under low stringency, medium stringency, high stringency, or very high stringency conditions” describes conditions for hybridization and washing.
- Guidance for performing hybridization reactions can be found in Ausubel et al ., (1998, supra ), Sections 6.3.1-6.3.6. Aqueous and non-aqueous methods are described in that reference and either can be used.
- Reference herein to low stringency conditions include and encompass from at least about 1% v/v to at least about 15% v/v formamide and from at least about 1 M to at least about 2 M salt for hybridization at 42° C, and at least about 1 M to at least about 2 M salt for washing at 42° C.
- Low stringency conditions also may include 1% Bovine Serum Albumin (BSA), 1 mM EDTA, 0.5 M NaHPO 4 (pH 7.2), 7% SDS for hybridization at 65°C, and (i) 2 ⁇ SSC, 0.1% SDS; or (ii) 0.5% BSA, 1 mM EDTA, 40 mM NaHPO 4 (pH 7.2), 5% SDS for washing at room temperature.
- BSA Bovine Serum Albumin
- One embodiment of low stringency conditions includes hybridization in 6 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2 ⁇ SSC, 0.1% SDS at least at 50° C (the temperature of the washes can be increased to 55° C for low stringency conditions).
- SSC sodium chloride/sodium citrate
- Medium stringency conditions include and encompass from at least about 16% v/v to at least about 30% v/v formamide and from at least about 0.5 M to at least about 0.9 M salt for hybridization at 42° C, and at least about 0.1 M to at least about 0.2 M salt for washing at 55° C.
- Medium stringency conditions also may include 1% Bovine Serum Albumin (BSA), 1 mM EDTA, 0.5 M NaHPO 4 (pH 7.2), 7% SDS for hybridization at 65° C, and (i) 2 ⁇ SSC, 0.1% SDS; or (ii) 0.5% BSA, 1 mM EDTA, 40 mM NaHPO 4 (pH 7.2), 5% SDS for washing at 60-65° C.
- BSA Bovine Serum Albumin
- medium stringency conditions includes hybridizing in 6 x SSC at about 45°C, followed by one or more washes in 0.2 ⁇ SSC, 0.1% SDS at 60°C.
- High stringency conditions include and encompass from at least about 31% v/v to at least about 50% v/v formamide and from about 0.01 M to about 0.15 M salt for hybridization at 42° C, and about 0.01 M to about 0.02 M salt for washing at 55° C.
- High stringency conditions also may include 1% BSA, 1 mM EDTA, 0.5 M NaHPO 4 (pH 7.2), 7% SDS for hybridization at 65° C, and (i) 0.2 ⁇ SSC, 0.1% SDS; or (ii) 0.5% BSA, 1 mM EDTA, 40 mM NaHPO 4 (pH 7.2), 1% SDS for washing at a temperature in excess of 65° C.
- One embodiment of high stringency conditions includes hybridizing in 6 ⁇ SSC at about 45°C, followed by one or more washes in 0.2 ⁇ SSC, 0.1% SDS at 65° C.
- One embodiment of very high stringency conditions includes hybridizing in 0.5 M sodium phosphate, 7% SDS at 65° C, followed by one or more washes in 0.2 ⁇ SSC, 1% SDS at 65° C.
- T m 81.5 + 16.6 (log 10 M) + 0.41 (%G+C) - 0.63 (% formamide) - (600/length)
- M is the concentration of Na + , preferably in the range of 0.01 molar to 0.4 molar
- %G+C is the sum of guanosine and cytosine bases as a percentage of the total number of bases, within the range between 30% and 75% G+C
- % formamide is the percent formamide concentration by volume
- length is the number of base pairs in the DNA duplex.
- the T m of a duplex DNA decreases by approximately 1° C with every increase of 1% in the number of randomly mismatched base pairs. Washing is generally carried out at T m - 15° C for high stringency, or T m - 30° C for moderate stringency.
- a membrane e.g ., a nitrocellulose membrane or a nylon membrane
- immobilized DNA is hybridized overnight at 42° C in a hybridization buffer (50% deionized formamide, 5 ⁇ SSC, 5 ⁇ Denhardt's solution (0.1% ficoll, 0.1% polyvinylpyrollidone and 0.1 % bovine serum albumin), 0.1% SDS and 200 mg/mL denatured salmon sperm DNA) containing a labeled probe.
- a hybridization buffer 50% deionized formamide, 5 ⁇ SSC, 5 ⁇ Denhardt's solution (0.1% ficoll, 0.1% polyvinylpyrollidone and 0.1 % bovine serum albumin), 0.1% SDS and 200 mg/mL denatured salmon sperm DNA
- the membrane is then subjected to two sequential medium stringency washes (i.e ., 2 ⁇ SSC, 0.1% SDS for 15 min at 45° C, followed by 2 ⁇ SSC, 0.1% SDS for 15 min at 50° C), followed by two sequential higher stringency washes ( i.e ., 0.2 ⁇ SSC, 0.1% SDS for 12 min at 55° C followed by 0.2 ⁇ SSC and 0.1% SDS solution for 12 min at 65-68° C.
- two sequential medium stringency washes i.e ., 2 ⁇ SSC, 0.1% SDS for 15 min at 45° C, followed by 2 ⁇ SSC, 0.1% SDS for 15 min at 50° C
- two sequential higher stringency washes i.e ., 0.2 ⁇ SSC, 0.1% SDS for 12 min at 55° C followed by 0.2 ⁇ SSC and 0.1% SDS solution for 12 min at 65-68° C.
- ArgRS polynucleotides that encode an ArgRS polypeptide.
- these embodiments may be utilized to recombinantly produce a desired ArgRS polypeptide or variant thereof, or to express the ArgRS polypeptide in a selected cell or subject.
- nucleotide sequences that encode a polypeptide as described herein.
- Some of these polynucleotides may bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention, for example polynucleotides that are optimized for human and/or primate codon selection.
- polynucleotides can encode the ArgRS polypeptides of the invention.
- polynucleotide sequence can be manipulated for various reasons. Examples include but are not limited to the incorporation of preferred codons to enhance the expression of the polynucleotide in various organisms (see generally Nakamura et al., Nuc. Acid. Res. (2000) 28 (1): 292 ).
- silent mutations can be incorporated in order to introduce, or eliminate restriction sites, decrease the density of CpG dinucleotide motifs (see for example, Kameda et al., Biochem. Biophys. Res. Commun.
- mammalian expression can be further optimized by including a Kozak consensus sequence [i.e., (a/g)cc(a/g)ccATGg] at the start codon.
- Kozak consensus sequences useful for this purpose are known in the art ( Mantyh et al. PNAS 92: 2662-2666 (1995 ); Mantyh et al. Prot. Exp. & Purif. 6,124 (1995 )).
- polynucleotides of the present invention may be combined with other DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a polynucleotide fragment of almost any length may be employed; with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol.
- Polynucleotides and fusions thereof may be prepared, manipulated and/or expressed using any of a variety of well-established techniques known and available in the art.
- polynucleotide sequences which encode polypeptides of the invention, or fusion proteins or functional equivalents thereof may be used in recombinant DNA molecules to direct expression of an ArgRS polypeptide in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences that encode substantially the same or a functionally equivalent amino acid sequence may be produced and these sequences may be used to clone and express a given polypeptide.
- codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce a recombinant RNA transcript having desirable properties, such as a half-life which is longer than that of a transcript generated from the naturally occurring sequence.
- Such polynucleotides are commonly referred to as "codon-optimized.” Any of the polynucleotides described herein may be utilized in a codon-optimized form.
- a polynucleotide can be codon optimized for use in specific bacteria such as E. coli or yeast such as S. cerevisiae (see, e.g., Burgess-Brown et al., Proteins Expr Purif. 59:94-102, 2008 ; Ermolaeva MD (2001) Curr. Iss. Mol. Biol. 3 (4) 91-7 ; Welch et al., PLoS ONE 4(9): e7007 doi:10.1371/journal.pone.0007002 ).
- polynucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter polypeptide encoding sequences for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, expression and/or activity of the gene product.
- polynucleotides encoding polypeptides of the invention may be delivered to a subject in vivo , e.g ., using gene therapy techniques.
- Gene therapy refers generally to the transfer of heterologous nucleic acids to the certain cells, target cells, of a mammal, particularly a human, with a disorder or conditions for which such therapy is sought.
- the nucleic acid is introduced into the selected target cells in a manner such that the heterologous DNA is expressed and a therapeutic product encoded thereby is produced.
- viral vectors that can be utilized for gene therapy as taught herein include adenovirus, herpes virus, vaccinia, adeno-associated virus (AAV), or, preferably, an RNA virus such as a retrovirus.
- the retroviral vector is a derivative of a murine or avian retrovirus, or is a lentiviral vector.
- the preferred retroviral vector is a lentiviral vector.
- retroviral vectors in which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), SIV, BIV, HIV and Rous Sarcoma Virus (RSV).
- MoMuLV Moloney murine leukemia virus
- HaMuSV Harvey murine sarcoma virus
- MuMTV murine mammary tumor virus
- SIV BIV
- HIV Rous Sarcoma Virus
- a number of additional retroviral vectors can incorporate multiple genes. All of these vectors can transfer or incorporate a gene for a selectable marker so that transduced cells can be identified and generated.
- the vector may be made target specific.
- Retroviral vectors can be made target specific by inserting, for example, a polynucleotide encoding a protein (dimer). Illustrative targeting may be accomplished by using an antibody to target the retroviral vector. Those of skill in the art will know of, or can readily ascertain without undue experimentation, specific polynucleotide sequences which can be inserted into the retroviral genome to allow target specific delivery of the retroviral vector containing the zinc finger-nucleotide binding protein polynucleotide.
- helper cell lines that contain plasmids encoding all of the structural genes of the retrovirus under the control of regulatory sequences within the LTR. These plasmids are missing a nucleotide sequence which enables the packaging mechanism to recognize an RNA transcript for encapsulation.
- Helper cell lines which have deletions of the packaging signal include but are not limited to PSI.2, PA317 and PA12, for example. These cell lines produce empty virions, since no genome is packaged.
- the vector can be packaged and vector virion produced.
- the vector virions produced by this method can then be used to infect a tissue cell line, such as NIH 3T3 cells, to produce large quantities of chimeric retroviral virions.
- Non-viral delivery techniques for gene therapy can also be used including, for example, DNA-ligand complexes, adenovirus-ligand-DNA complexes, direct injection of DNA, CaPO 4 precipitation, gene gun techniques, electroporation, liposomes, lipofection, and the like. Any of these methods are widely available to one skilled in the art and would be suitable for use in the present invention. Other suitable methods are available to one skilled in the art, and it is to be understood that the present invention can be accomplished using any of the available methods of transfection. Lipofection can be accomplished by encapsulating an isolated DNA molecule within a liposomal particle and contacting the liposomal particle with the cell membrane of the target cell.
- Liposomes are self-assembling, colloidal particles in which a lipid bilayer, composed of amphiphilic molecules such as phosphatidyl serine or phosphatidyl choline, encapsulates a portion of the surrounding media such that the lipid bilayer surrounds a hydrophilic interior.
- Unilammellar or multilammellar liposomes can be constructed such that the interior contains a desired chemical, drug, or, as in the instant invention, an isolated DNA molecule.
- polynucleotides encoding polypeptides of the invention may be used to express and delivery an ArgRS polypeptide via cell therapy.
- the current invention includes a cell therapy for treating a disease or disorder, comprising administering a host cell expressing, or capable of expressing, an ArgRS polypeptide.
- Cell therapy involves the administration of cells which have been selected, multiplied and pharmacologically treated or altered (i.e. genetically modified) outside of the body ( Bordignon, C. et al, Cell Therapy: Achievements and Perspectives (1999), Haematologica, 84, pp.1110-1149 ).
- host cells include for example, primary cells, including macrophages, and stem cells which have been genetically modified to express an ArgRS polypeptide.
- the aim of cell therapy is to replace, repair or enhance the biological function of damaged tissues or organs.
- Transplanted cells may function by releasing bioactive compounds such as an ArgRS polypeptide of the invention, to replace endogenous ArgRS polypeptides which are absent or produced in insufficient quantities in an effected system.
- Embodiments of the present invention also include oligonucleotides, whether for detection, amplification, antisense therapies, or other purpose.
- oligonucleotide or “oligo” or “oligomer” is intended to encompass a singular "oligonucleotide” as well as plural “oligonucleotides,” and refers to any polymer of two or more of nucleotides, nucleosides, nucleobases or related compounds used as a reagent in the amplification methods of the present invention, as well as subsequent detection methods.
- the oligonucleotide may be DNA and/or RNA and/or analogs thereof.
- oligonucleotide does not necessarily denote any particular function to the reagent, rather, it is used generically to cover all such reagents described herein.
- An oligonucleotide may serve various different functions, e.g ., it may function as a primer if it is capable of hybridizing to a complementary strand and can further be extended in the presence of a nucleic acid polymerase, it may provide a promoter if it contains a sequence recognized by an RNA polymerase and allows for transcription, and it may function to prevent hybridization or impede primer extension if appropriately situated and/or modified.
- An oligonucleotide may also function as a probe, or an antisense agent.
- An oligonucleotide can be virtually any length, limited only by its specific function, e.g ., in an amplification reaction, in detecting an amplification product of the amplification reaction, or in an antisense or RNA interference application. Any of the oligonucleotides described herein can be used as a primer, a probe, an antisense oligomer, or an RNA interference agent.
- primer refers to a single-stranded oligonucleotide capable of acting as a point of initiation for template-directed DNA synthesis under suitable conditions defined, for example, by buffer and temperature, in the presence of four different nucleoside triphosphates and an agent for polymerization, such as a DNA or RNA polymerase or reverse transcriptase.
- the length of the primer in any given case, depends on, for example, the intended use of the primer, and generally ranges from about 15 to 30 nucleotides, although shorter and longer primers may be used. Short primer molecules generally require cooler temperatures to form sufficiently stable hybrid complexes with the template.
- a primer need not reflect the exact sequence of the template but must be sufficiently complementary to hybridize with such template.
- the primer site is the area of the template to which a primer hybridizes.
- the primer pair is a set of primers including a 5' upstream primer that hybridizes with the 5' end of the sequence to be amplified and a 3' downstream primer that hybridizes with the complement of the 3' end of the sequence to be amplified.
- probe as used herein includes a surface-immobilized or soluble but capable of being immobilized molecule that can be recognized by a particular target. See, e.g., U.S. Patent No. 6,582,908 for an example of arrays having all possible combinations of probes with 10, 12, and more bases.
- Probes and primers as used herein typically comprise at least 10-15 contiguous nucleotides of a known sequence. In order to enhance specificity, longer probes and primers may also be employed, such as probes and primers that comprise at least 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, or at least 150 nucleotides of an ArgRS reference sequence or its complement. Probes and primers may be considerably longer than these examples, and it is understood that any length supported by the knowledge in the art and the specification, including the tables, figures, and Sequence Listing, may be used.
- PCR primer pairs can be derived from a known sequence, for example, by using computer programs intended for that purpose such as Primer (Version 0.5, 1991, Whitehead Institute for Biomedical Research, Cambridge Mass.).
- Oligonucleotides for use as primers or probes may be selected using software known in the art. For example, OLIGO 4.06 software is useful for the selection of PCR primer pairs of up to 100 nucleotides each, and for the analysis of oligonucleotides and larger polynucleotides of up to 5,000 nucleotides from an input polynucleotide sequence of up to 32 kilobases. Similar primer selection programs have incorporated additional features for expanded capabilities. For example, the PrimOU primer selection program (available to the public from the Genome Center at University of Texas South West Medical Center, Dallas Tex.) is capable of choosing specific primers from megabase sequences and is thus useful for designing primers on a genome-wide scope.
- the Primer3 primer selection program (available to the public from the Whitehead Institute/MIT Center for Genome Research, Cambridge Mass.) allows the user to input a "mispriming library," in which sequences to avoid as primer binding sites are user-specified. Primer3 is useful, in particular, for the selection of oligonucleotides for microarrays. (The source code for the latter two primer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs.)
- the PrimeGen program (available to the public from the UK Human Genome Mapping Project Resource Centre, Cambridge UK) designs primers based on multiple sequence alignments, thereby allowing selection of primers that hybridize to either the most conserved or least conserved regions of aligned nucleic acid sequences.
- this program is useful for identification of both unique and conserved oligonucleotides and polynucleotide fragments.
- the oligonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing primers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids. Methods of oligonucleotide selection are not limited to those described herein.
- oligonucleotides can be prepared by stepwise solid-phase synthesis, employing methods detailed in the references cited above, and below with respect to the synthesis of oligonucleotides having a mixture or uncharged and cationic backbone linkages.
- additional chemical moieties to the oligonucleotide, e.g ., to enhance pharmacokinetics or to facilitate capture or detection of the compound.
- Such a moiety may be covalently attached, typically to a terminus of the oligomer, according to standard synthetic methods.
- addition of a polyethyleneglycol moiety or other hydrophilic polymer may be useful in enhancing solubility.
- One or more charged groups e.g ., anionic charged groups such as an organic acid, may enhance cell uptake.
- detectable molecules may be used to render an oligonucleotide, or protein detectable, such as a radioisotopes, fluorochromes, dyes, enzymes, nanoparticles, chemiluminescent markers, biotin, or other monomer known in the art that can be detected directly ( e.g ., by light emission) or indirectly ( e.g ., by binding of a fluorescently-labeled antibody).
- a radioisotopes e.g ., fluorochromes, dyes, enzymes, nanoparticles, chemiluminescent markers, biotin, or other monomer known in the art that can be detected directly ( e.g ., by light emission) or indirectly ( e.g ., by binding of a fluorescently-labeled antibody).
- Radioisotopes provide examples of detectable molecules that can be utilized in certain aspects of the present invention.
- Several radioisotopes can be used as detectable molecules for labeling nucleotides or proteins, including, for example, 32 P, 33 P, 35 S, 3 H, and 125 I. These radioisotopes have different half-lives, types of decay, and levels of energy which can be tailored to match the needs of a particular protocol.
- 3 H is a low energy emitter which results in low background levels, however this low energy also results in long time periods for autoradiography.
- Radioactively labeled ribonucleotides, deoxyribonucleotides and amino acids are commercially available.
- Nucleotides are available that are radioactively labeled at the first, or ⁇ , phosphate group, or the third, or ⁇ , phosphate group.
- both [ ⁇ - 32 P] dATP and [ ⁇ - 32 P] dATP are commercially available.
- different specific activities for radioactively labeled nucleotides are also available commercially and can be tailored for different protocols.
- fluorophores can be used for labeling nucleotides including, for example, fluorescein, tetramethylrhodamine, Texas Red, and a number of others ( e.g ., Haugland, Handbook of Fluorescent Probes - 9th Ed., 2002, Molec. Probes, Inc., Eugene OR; Haugland, The Handbook: A Guide to Fluorescent Probes and Labeling Technologies-10th Ed., 2005, Invitrogen, Carlsbad, CA ).
- oligonucleotides may be fluorescently labeled during chemical synthesis, since incorporation of amines or thiols during nucleotide synthesis permit addition of fluorophores.
- Fluorescently labeled nucleotides are commercially available.
- uridine and deoxyuridine triphosphates are available that are conjugated to ten different fluorophores that cover the spectrum.
- Fluorescent dyes that can be bound directly to nucleotides can also be utilized as detectable molecules.
- FAM, JOE, TAMRA, and ROX are amine reactive fluorescent dyes that have been attached to nucleotides and are used in automated DNA sequencing.
- These fluorescently labeled nucleotides for example, ROX-ddATP, ROX-ddCTP, ROX-ddGTP and ROX-ddUTP, are commercially available.
- Non-radioactive and non-fluorescent detectable molecules are also available.
- biotin can be attached directly to nucleotides and detected by specific and high affinity binding to avidin or streptavidin which has been chemically coupled to an enzyme catalyzing a colorimetric reaction (such as phosphatase, luciferase, or peroxidase).
- Digoxigenin labeled nucleotides can also similarly be used for non-isotopic detection of nucleic acids. Biotinylated and digoxigenin-labeled nucleotides are commercially available.
- Nanoparticles also can be used to label oligonucleotide probes. These particles range from 1-1000 nm in size and include diverse chemical structures such as gold and silver particles and quantum dots. When irradiated with angled incident white light, silver or gold nanoparticles ranging from 40-120 nm will scatter monochromatic light with high intensity. The wavelength of the scattered light is dependent on the size of the particle. Four to five different particles in close proximity will each scatter monochromatic light, which when superimposed will give a specific, unique color. The particles are being manufactured by companies such as Genicon Sciences (Carlsbad, CA).
- Derivatized silver or gold particles can be attached to a broad array of molecules including, proteins, antibodies, small molecules, receptor ligands, and nucleic acids.
- the surface of the particle can be chemically derivatized to allow attachment to a nucleotide.
- Quantum dots are fluorescing crystals 1-5 nm in diameter that are excitable by light over a large range of wavelengths. Upon excitation by light having an appropriate wavelength, these crystals emit light, such as monochromatic light, with a wavelength dependent on their chemical composition and size. Quantum dots such as CdSe, ZnSe, InP, or InAs possess unique optical properties; these and similar quantum dots are available from a number of commercial sources ( e.g ., NN-Labs, Fayetteville, AR; Ocean Nanotech, Fayetteville, AR; Nanoco Technologies, Manchester, UK; Sigma-Aldrich, St. Louis, MO).
- the size classes of the crystals are created either 1) by tight control of crystal formation parameters to create each desired size class of particle, or 2) by creation of batches of crystals under loosely controlled crystal formation parameters, followed by sorting according to desired size and/or emission wavelengths.
- Two examples of references in which quantum dots are embedded within intrinsic silicon epitaxial layers of semiconductor light emitting/detecting devices are United States Patent Nos. 5,293,050 and 5,354,707 to Chapple Sokol, et al.
- oligonucleotide primers or probes may be labeled with one or more light-emitting or otherwise detectable dyes.
- the light emitted by the dyes can be visible light or invisible light, such as ultraviolet or infrared light.
- the dye may be a fluorescence resonance energy transfer (FRET) dye; a xanthene dye, such as fluorescein and rhodamine; a dye that has an amino group in the alpha or beta position (such as a naphthylamine dye, 1-dimethylaminonaphthyl-5-sulfonate, 1-anilino-8-naphthalende sulfonate and 2-p-touidinyl-6-naphthalene sulfonate); a dye that has 3-phenyl-7-isocyanatocoumarin; an acridine, such as 9-isothiocyanatoacridine and acridine orange; a pyrene, a bensoxadiazole and a stilbene; a dye that has 3-( ⁇ -carboxypentyl)-3'-ethyl-5,5'-dimethyloxacarbocyanine (CYA); 6-carboxy
- FRET flu
- ArgRS polynucleotides and oligonucleotides of the present invention can be used in any of the therapeutic, diagnostic, research, or drug discovery compositions and methods described herein.
- the present invention further provides antibodies that exhibit binding specificity for an ArgRS polypeptide, or its native cellular binding partner (i.e. cellular receptor, lipid, carbohydrate, protein, or nucleic acid binding partner), or complex thereof, and methods of using the same.
- the term antibody includes the various variations of the same, such as FABs, human antibodies, modified human antibodies, single chains, nonhuman antibodies, and other derivatives of the immunoglobulin fold that underlie immune system ligands for antigens, as described herein and known in the art.
- Antibodies can be used in any of the therapeutic, diagnostic, drug discovery, or protein expression/purification methods and compositions provided herein.
- Certain antibodies of the present invention differ from certain previously made antibodies because they can distinguish between the ArgRS protein fragments of SEQ ID NO: 29, 151, or 137 and their corresponding full-length ArgRS, typically by binding with greater affinity to the ArgRS protein fragments than to the corresponding full-length ArgRS.
- Such antibodies may bind to unique sequences or structures generated or revealed by splice variations, proteolysis, or other cellular processing that generates an ArgRS protein fragment of the invention (e.g ., post translational processing, including but not limited to phosphorylation and other modifications that change protein structure).
- such antibodies may have binding specificity to one or more non-solvent exposed faces that are exposed in the ArgRS protein fragment but not in the full-length ArgRS, or sequences that are not found or are otherwise inaccessible in the full-length ArgRS.
- Antibodies may also bind to unique three-dimensional structures that result from differences in folding between the ArgRS protein fragment and the full-length ArgRS. Such differences in folding may be localized ( e.g., to a specific domain or region) or globalized. As one example, folding of ArgRS protein fragments may generate unique continuous or discontinuous epitopes that are not found in the corresponding or parent ArgRS.
- Examples also include antibodies that specifically bind to N- or C- termini generated by splice variations, proteolysis, or other cellular processing; such termini may be unique compared to the full-length ArgRS or may not be exposed for antibody binding in the full-length versions due to their termini being completely or partially buried in the overall structure of the larger ArgRS parent molecule.
- antibodies provided herein do not form aggregates, have a desired solubility, and/or have an immunogenicity profile that is suitable for use in humans, as described herein and known in the art. Also included are antibodies that are suitable for production work, such as to purify the ArgRS protein fragments described herein. Preferably, active antibodies can be concentrated to at least about 10mg/ml and optional formulated for biotherapeutic uses.
- antibodies are effective for modulating one or more of the non-canonical activities mediated by an ArgRS polypeptide of the invention.
- the antibody is one that binds to an ArgRS polypeptide and/or its binding partner, inhibits their ability to interact with each other, and/or antagonizes the non-canonical activity of the ArgRS polypeptide.
- the antibody binds to the cellular binding partner of an ArgRS polypeptide, and mimics the ArgRS polypeptide activity, such as by increasing or agonizing the non-canonical activity mediated by the ArgRS polypeptide. Accordingly, antibodies may be used to diagnose, treat, or prevent diseases, disorders or other conditions that are mediated by an ArgRS polypeptide of the invention, such as by antagonizing or agonizing its activity partially or fully.
- an antibody, or antigen-binding fragment thereof is said to "specifically bind,” “immunologically bind,” and/or is “immunologically reactive” to a polypeptide of the invention if it reacts at a detectable level (within, for example, an ELISA assay) with the polypeptide, and does not react detectably in a statistically significant manner with unrelated polypeptides under similar conditions.
- a binding agent does not significantly interact with a full-length version of the ArgRS polypeptide.
- Immunological binding generally refers to the non-covalent interactions of the type which occur between an immunoglobulin molecule and an antigen for which the immunoglobulin is specific.
- the strength, or affinity of binding such as immunological binding interactions can be expressed in terms of the dissociation constant (K d ) of the interaction, wherein a smaller K d represents a greater affinity.
- Immunological binding properties of selected polypeptides can be quantified using methods well known in the art. See, e.g., Davies et al. (1990) Annual Rev. Biochem. 59:439-473 .
- an antibody has an affinity for an ArgRS protein fragment of at least about 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, or 50 nM.
- the affinity of the antibody for an ArgRS protein fragment is stronger than its affinity for a corresponding full-length ArgRS polypeptide, typically by about 1.5x, 2x, 2.5x, 3x, 3.5x, 4x, 4.5x, 5x, 6x, 7x, 8x, 9x, 10x, 15x, 20x, 25x, 30x, 40x, 50x, 60x, 70x, 80x, 90x, 100x, 200x, 300x, 400x, 500x, 600x, 700x, 800x, 900x, 1000x or more (including all integers in between).
- an antibody as an affinity for a corresponding full-length ArgRS protein of at least about 0.05, 0.1, 0.25, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 ⁇ M. In certain embodiments, an antibody binds weakly or substantially undetectably to a full-length ArgRS protein.
- an “antigen-binding site,” or “binding portion” of an antibody refers to the part of the immunoglobulin molecule that participates in antigen binding.
- the antigen binding site is formed by amino acid residues of the N-terminal variable ("V") regions of the heavy ("H") and light (“L”) chains.
- V N-terminal variable
- H heavy
- L light
- Three highly divergent stretches within the V regions of the heavy and light chains are referred to as “hypervariable regions" which are interposed between more conserved flanking stretches known as “framework regions,” or "FRs".
- FR refers to amino acid sequences which are naturally found between and adjacent to hypervariable regions in immunoglobulins.
- the three hypervariable regions of a light chain and the three hypervariable regions of a heavy chain are disposed relative to each other in three dimensional space to form an antigen-binding surface.
- the antigen-binding surface is complementary to the three-dimensional surface of a bound antigen, and the three hypervariable regions of each of the heavy and light chains are referred to as "complementarity-determining regions,” or "CDRs.”
- Antibodies may be prepared by any of a variety of techniques known to those of ordinary skill in the art. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988 . Monoclonal antibodies specific for a polypeptide of interest may be prepared, for example, using the technique of Kohler and Milstein, Eur. J. Immunol. 6:511-519, 1976 , and improvements thereto. Also included are methods that utilize transgenic animals such as mice to express human antibodies.
- the polypeptides of this invention may be used in the purification process in, for example, an affinity chromatography step.
- Fv fragment can be produced by preferential proteolytic cleavage of an IgM, and on rare occasions IgG or IgA immunoglobulin molecule. Fv fragments are, however, more commonly derived using recombinant techniques known in the art.
- the Fv fragment includes a non-covalent V H ::V L heterodimer including an antigen-binding site which retains much of the antigen recognition and binding capabilities of the native antibody molecule. See, e.g., Inbar et al. (1972) Proc. Nat. Acad. Sci. USA 69:2659-2662 ; Hochman et al. (1976) Biochem 15:2706-2710 ; and Ehrlich et al. (1980) Biochem 19:4091-4096 .
- a single chain Fv (“sFv”) polypeptide is a covalently linked V H ::V L heterodimer which is expressed from a gene fusion including V H - and V L -encoding genes linked by a peptide-encoding linker.
- a number of methods have been described to discern chemical structures for converting the naturally aggregated--but chemically separated--light and heavy polypeptide chains from an antibody V region into an sFv molecule which will fold into a three dimensional structure substantially similar to the structure of an antigen-binding site. See, e.g., U.S. Pat. Nos. 5,091,513 and 5,132,405, to Huston et al .; and U.S. Pat. No. 4,946,778, to Ladner et al.
- Each of the above-described molecules includes a heavy chain and a light chain CDR set, respectively interposed between a heavy chain and a light chain FR set which provide support to the CDRS and define the spatial relationship of the CDRs relative to each other.
- CDR set refers to the three hypervariable regions of a heavy or light chain V region. Proceeding from the N-terminus of a heavy or light chain, these regions are denoted as "CDR1," "CDR2,” and “CDR3" respectively.
- An antigen-binding site therefore, includes six CDRs, comprising the CDR set from each of a heavy and a light chain V region.
- a polypeptide comprising a single CDR (e.g ., a CDR1, CDR2 or CDR3) is referred to herein as a "molecular recognition unit.” Crystallographic analysis of a number of antigen-antibody complexes has demonstrated that the amino acid residues of CDRs form extensive contact with bound antigen, wherein the most extensive antigen contact is with the heavy chain CDR3. Thus, the molecular recognition units are primarily responsible for the specificity of an antigen-binding site.
- FR set refers to the four flanking amino acid sequences which frame the CDRs of a CDR set of a heavy or light chain V region. Some FR residues may contact bound antigen; however, FRs are primarily responsible for folding the V region into the antigen-binding site, particularly the FR residues directly adjacent to the CDRS. Within FRs, certain amino residues and certain structural features are very highly conserved. In this regard, all V region sequences contain an internal disulfide loop of around 90 amino acid residues. When the V regions fold into a binding-site, the CDRs are displayed as projecting loop motifs which form an antigen-binding surface.
- sdAbs or “nanobodies” refer to an antibody fragment consisting of a single monomeric variable antibody domain (see, e.g., U.S. Patent Nos. 5,840,526 ; 5,874,541 ; 6,005,079 , 6,765,087 , 5,800,988 ; 5,874,541 ; and 6,015,695 ).
- sdABs typically have a molecular weight of about 12-15 kDa.
- sdABs and other antibody molecules can be derived or isolated from the unique heavy-chain antibodies of immunized camels and llamas, often referred to as camelids. See, e.g., Conrath et al., JBC. 276:7346-7350, 2001 .
- the antibodies of the present invention can be used in any of the therapeutic, diagnostic, drug discovery, protein purification, and analytical methods and compositions described herein.
- the present invention further provides antibody alternatives or other binding agents, such as soluble receptors, adnectins, peptides, peptide mimetics, small molecules, aptamers, etc., that exhibit binding specificity for an ArgRS polypeptide or its cellular binding partner as disclosed herein, or to a portion, variant or derivative thereof, and compositions and methods of using same.
- Binding agents can be used in any of the therapeutic, diagnostic, drug discovery, or protein expression/purification, and analytical methods and compositions described herein.
- Biologic-based binding agents such as adnectins, soluble receptors, avimers, and trinectins are particularly useful.
- such binding agents are effective for modulating one or more of the non-canonical activities mediated by an ArgRS polypeptide of the invention.
- the binding agent is one that binds to an ArgRS polypeptide and/or its binding partner, inhibits their ability to interact with each other, and/or antagonizes the non-canonical activity of the ArgRS polypeptide.
- the binding agent binds to the cellular binding partner of an ArgRS polypeptide, and mimics the ArgRS polypeptide activity, such as by increasing or agonizing the non-canonical activity mediated by the ArgRS polypeptide. Accordingly, such binding agents may be used to diagnose, treat, or prevent diseases, disorders or other conditions that are mediated by an ArgRS polypeptide of the invention, such as by antagonizing or agonizing its activity partially or fully.
- a binding agent is said to "specifically bind" to an ArgRS polypeptide of the invention, or its cellular binding partner, if it reacts at a detectable level (within, for example, an ELISA assay) with the polypeptide or its cellular binding partner, and does not react detectably in a statistically significant manner with unrelated polypeptides under similar conditions. In certain instances, a binding agent does not significantly interact with a full-length version of the ArgRS polypeptide.
- a binding agent has an affinity for an ArgRS protein fragment or its cellular binding partner of at least about 0.01, 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, or 50 nM.
- the affinity of the binding agent for an ArgRS protein fragment is stronger than its affinity for a corresponding full-length ArgRS polypeptide, typically by about 1.5x, 2x, 2.5x, 3x, 3.5x, 4x, 4.5x, 5x, 6x, 7x, 8x, 9x, 10x, 15x, 20x, 25x, 30x, 40x, 50x, 60x, 70x, 80x, 90x, 100x, 200x, 300x, 400x, 500x, 600x, 700x, 800x, 900x, 1000x or more (including all integers in between).
- a binding agent has an affinity for a corresponding full-length ArgRS protein of at least about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 ⁇ M.
- peptides are included as binding agents.
- the term peptide typically refers to a polymer of amino acid residues and to variants and synthetic analogues of the same.
- the term “peptide” refers to relatively short polypeptides, including peptides that consist of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, or 50 amino acids, including all integers and ranges ( e.g., 5-10, 8-12, 10-15) in between, and interact with an ArgRS polypeptide, its cellular binding partner, or both.
- Peptides can be composed of naturally-occurring amino acids and/or non-naturally occurring amino acids, as described herein.
- peptidomimetics or peptide analogs are also provided.
- Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of non-peptide compound are termed "peptide mimetics” or “peptidomimetics” ( Luthman, et al., A Textbook of Drug Design and Development, 14:386-406, 2nd Ed., Harwood Academic Publishers (1996 ); Joachim Grante, Angew. Chem. Int. Ed. Engl., 33:1699-1720 (1994 ); Fauchere, J., Adv.
- a peptidomimetic is a molecule that mimics the biological activity of a peptide but is no longer peptidic in chemical nature. Peptidomimetic compounds are known in the art and are described, for example, in U.S. Patent No. 6,245,886 .
- the present invention also includes peptoids.
- Peptoid derivatives of peptides represent another form of modified peptides that retain the important structural determinants for biological activity, yet eliminate the peptide bonds, thereby conferring resistance to proteolysis ( Simon, et al., PNAS USA. 89:9367-9371, 1992 ).
- Peptoids are oligomers of N-substituted glycines. A number of N-alkyl groups have been described, each corresponding to the side chain of a natural amino acid.
- the peptidomimetics of the present invention include compounds in which at least one amino acid, a few amino acids or all amino acid residues are replaced by the corresponding N-substituted glycines.
- Peptoid libraries are described, for example, in U.S. Patent No. 5,811,387 .
- a binding agent may also include one or more small molecules.
- a "small molecule” refers to an organic compound that is of synthetic or biological origin (biomolecule), but is typically not a polymer.
- Organic compounds refer to a large class of chemical compounds whose molecules contain carbon, typically excluding those that contain only carbonates, simple oxides of carbon, or cyanides.
- a “biomolecule” refers generally to an organic molecule that is produced by a living organism, including large polymeric molecules (biopolymers) such as peptides, polysaccharides, and nucleic acids as well, and small molecules such as primary secondary metabolites, lipids, phospholipids, glycolipids, sterols, glycerolipids, vitamins, and hormones.
- a “polymer” refers generally to a large molecule or macromolecule composed of repeating structural units, which are typically connected by covalent chemical bond.
- a small molecule has a molecular weight of less than 1000-2000 Daltons, typically between about 300 and 700 Daltons, and including about 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 500, 650, 600, 750, 700, 850, 800, 950, 1000 or 2000 Daltons. Small molecule libraries are described elsewhere herein.
- Aptamers are also included as binding agents (see, e.g., Ellington et al., Nature. 346, 818-22, 1990 ; and Tuerk et al., Science. 249, 505-10, 1990 ).
- Examples of aptamers included nucleic acid aptamers (e.g., DNA aptamers, RNA aptamers) and peptide aptamers.
- Nucleic acid aptamers refer generally to nucleic acid species that have been engineered through repeated rounds of in vitro selection or equivalent method, such as SELEX (systematic evolution of ligands by exponential enrichment), to bind to various molecular targets such as small molecules, proteins, nucleic acids, and even cells, tissues and organisms.
- nucleic acid aptamers that bind to the ArgRS polypeptides described herein and/or their cellular binding partners.
- Peptide aptamers typically include a variable peptide loop attached at both ends to a protein scaffold, a double structural constraint that typically increases the binding affinity of the peptide aptamer to levels comparable to that of an antibody's ( e.g., in the nanomolar range).
- the variable loop length may be composed of about 10-20 amino acids (including all integers in between), and the scaffold may include any protein that has good solubility and compacity properties.
- Certain exemplary embodiments may utilize the bacterial protein Thioredoxin-A as a scaffold protein, the variable loop being inserted within the reducing active site (-Cys-Gly-Pro-Cys- loop in the wild protein), with the two cysteines lateral chains being able to form a disulfide bridge.
- Methods for identifying peptide aptamers are described, for example, in U.S. Application No. 2003/0108532 .
- peptide aptamers that bind to the ArgRS polypeptides described herein and/or their cellular binding partners.
- Peptide aptamer selection can be performed using different systems known in the art, including the yeast two-hybrid system.
- ADNECTINSTM refers to a class of targeted biologies derived from human fibronectin, an abundant extracellular protein that naturally binds to other proteins. See, e.g., U.S. Application Nos. 2007/0082365 ; 2008/0139791 ; and 2008/0220049 .
- ADNECTINSTM typically consists of a natural fibronectin backbone, as well as the multiple targeting domains of a specific portion of human fibronectin. The targeting domains can be engineered to enable an ADNECTINTM to specifically recognize a therapeutic target of interest, such as an ArgRS protein fragment of the invention.
- AVIMERSTM refer to multimeric binding proteins or peptides engineered using in vitro exon shuffling and phage display. Multiple binding domains are linked, resulting in greater affinity and specificity compared to single epitope immunoglobulin domains. See, e.g., Silverman et al., Nature Biotechnology. 23:1556-1561, 2005 ; U.S. Patent No. 7,166,697 ; and U.S. Application Nos. 2004/0175756 , 2005/0048512 , 2005/0053973 , 2005/0089932 and 2005/0221384 .
- DARPins ankyrin repeat proteins
- DARPins are designed ankyrin repeat proteins that can offer advantages over antibodies for target binding in drug discovery and drug development.
- DARPins are ideally suited for in vivo imaging or delivery of toxins or other therapeutic payloads because of their favorable molecular properties, including small size and high stability.
- the low-cost production in bacteria and the rapid generation of many target-specific DARPins make the DARPin approach useful for drug discovery.
- DARPins can be easily generated in multispecific formats, offering the potential to target an effector DARPin to a specific organ or to target multiple receptors with one molecule composed of several DARPins. See, e.g., Stumpp et al., Curr Opin Drug Discov Devel. 10:153-159, 2007 ; U.S. Application No. 2009/0082274 ; and PCT/EP2001/10454 .
- FNfn10 10th fibronectin type III domain of human fibronectin
- FNfn10 10th fibronectin type III domain of human fibronectin
- the FNfn10 scaffold is compatible with virtually any display technologies. See, e.g., Batori et al., Protein Eng. 15:1015-20, 2002 ; and Wojcik et al., Nat Struct Mol Biol., 2010 ; and U.S. Patent No. 6,673,901 .
- Anticalins refer to a class of antibody mimetics, which are typically synthesized from human lipocalins, a family of binding proteins with a hypervariable loop region supported by a structurally rigid framework. See, e.g., U.S. Application No. 2006/0058510 . Anticalins typically have a size of about 20 kDa. Anticalins can be characterized by a barrel structure formed by eight antiparallel ⁇ -strands (a stable ⁇ -barrel scaffold) that are pairwise connected by four peptide loops and an attached ⁇ -helix. In certain aspects, conformational deviations to achieve specific binding are made in the hypervariable loop region(s). See, e.g., Skerra, FEBS J. 275:2677-83, 2008 .
- bioassays that relate to the ArgRS protein fragments and related agents as therapeutic and diagnostic reagents. Examples include bioassays and analytical assays that measure purity, biological activity, affinity, solubility, pH, endotoxin levels, among others, many of which are described herein. Also included are assays that establish dose response curves and/or provide one or more bases for comparison between different batches of agents. Batch comparisons can be based on any one or more of chemical characterization, biological characterization, and clinical characterization. For protein agents, also included are methods of evaluating the potency, stability, pharmacokinetics, and immunogenicity of a selected agent. Among other uses, these and other methods can be used for lot releasing testing of biologic or chemical agents, including the ArgRS protein fragments, antibodies, binding agents, polynucleotides such as antisense agents and vectors, and others described herein.
- bioaffinity assays Such assays can be used to assess the binding affinity, for example, between an ArgRS protein fragment and a cellular binding partner, or between an ArgRS protein fragment and an antibody. Binding affinity can also be measured between an ArgRS protein fragment and an alternate binding agent such as a candidate or lead test compound ( e.g ., small molecule modulator of an ArgRS), or between an ArgRS cellular binding partner and a candidate or lead test compound.
- a candidate or lead test compound e.g ., small molecule modulator of an ArgRS
- Certain exemplary binding affinity assays may utilize ELISA assays, as described herein and known in the art. Certain assays utilize high-performance receptor binding chromatography ( see, e.g., Roswall et al., Biologicals.
- binding affinity assays may utilize surface plasmon resonance (SPR)-based technologies.
- SPR surface plasmon resonance
- BIACore technologies certain of which integrate SPR technology with a microfluidics system to monitor molecular interactions in real time at concentrations ranging from pM to mM.
- KINEXATM assays also included are KINEXATM assays, which provide accurate measurements of binding specificity, binding affinity, and binding kinetics/rate constants.
- Certain embodiments relate to immunoassays for evaluating or optimizing the immunogenicity of protein agents.
- examples include ex vivo human cellular assays and in vitro immuno-enzymatic assays to provide useful information on the immunogenic potential of a therapeutic protein.
- Ex vivo cell-response assays can be used, for example, to reproduce the cellular co-operation between antigen-presenting cells (APCs) and T-cells, and thereby measure T-cells activation after contact with a protein of interest.
- APCs antigen-presenting cells
- Certain in vitro enzymatic assays may utilize a collection of recombinant HLA-DR molecules that cover a significant portion of a relevant human population, and may include automated immuno-enzymatic assays for testing the binding of peptides (stemming from the fragmentation of the therapeutic protein) with the HLA-DR molecules. Also included are methods of reducing the immunogenicity of a selected protein, such as by using these and related methods to identify and then remove or alter one or more T-cell epitopes from a protein agent.
- biological release assays e.g ., cell-based assays
- biological assays include, for example, cell-based assays that utilize a cellular binding partner (e.g ., cell-surface receptor) of a selected ArgRS protein fragment, which is functionally coupled to a readout, such as a fluorescent or luminescent indicator of a non-canonical biological activity, as described herein.
- a cellular binding partner e.g ., cell-surface receptor
- a readout such as a fluorescent or luminescent indicator of a non-canonical biological activity
- specific embodiments include a cell that comprises a cell-surface receptor or an extracellular portion thereof that binds to an ArgRS protein fragment, wherein the cell comprises a detector or readout.
- cytotoxicity-based biological assays include release assays (e.g ., chromium or europium release assays to measure apoptosis; see, e.g., von Zons et al., Clin Diagn Lab Immunol.4:202-207, 1997 ), among others, which can assess the cytotoxicity of ArgRS protein fragments, whether for establishing dose response curves, batch testing, or other properties related to approval by various regulatory agencies, such as the Food and Drug Administration (FDA).
- release assays e.g chromium or europium release assays to measure apoptosis; see, e.g., von Zons et al., Clin Diagn Lab Immunol.4:202-207, 1997
- FDA Food and Drug Administration
- a dose-response curve is an X-Y graph that relates the magnitude of a stressor to the response of a receptor; the response may be a physiological or biochemical response, such as a non-canonical biological activity in a cell in vitro or in a cell or tissue in vivo , a therapeutically effective amount as measured in vivo ( e.g., as measured by EC 50 ), or death, whether measured in vitro or in vivo ( e.g., cell death, organismal death).
- Death is usually indicated as an LD 50 , a statistically-derived dose that is lethal to 50% of a modeled population, though it can be indicated by LC 01 (lethal dose for 1% of the animal test population), LC 100 (lethal dose for 100% of the animal test population), or LC LO (lowest dose causing lethality). Almost any desired effect or endpoint can be characterized in this manner.
- the measured dose of a response curve is typically plotted on the X axis and the response is plotted on the Y axis. More typically, the logarithm of the dose is plotted on the X axis, most often generating a sigmoidal curve with the steepest portion in the middle.
- the No Observable Effect Level refers to the lowest experimental dose for which no measurable effect is observed, and the threshold dose refers to the first point along the graph that indicates a response above zero.
- NOEL No Observable Effect Level
- stronger drugs generate steeper dose response curves.
- the desired effects are found at doses slightly greater than the threshold dose, often because lower doses are relatively ineffective and higher doses lead to undesired side effects.
- a curve can be characterized by values such as ⁇ g/kg, mg/kg, or g/kg of body-weight, if desired.
- CV coefficient of variation
- different dose response curves of different batches e.g ., between different batches of ArgRS protein fragments, antibodies, or other agents
- CV allows comparison between data sets with different units or different means.
- two or three or more different batches of ArgRS protein fragments or other agents have a CV between them of less than about 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% for a 4, 5, 6, 7, or 8 point dose curve.
- the dose response curve is measured in a cell-based assay, and its readout relates to an increase or a decrease in a selected non-canonical activity of the ArgRS protein fragment.
- the dose response curve is measured in a cell release assay or animal model (e.g ., mouse model), and its readout relates to cell death or animal death.
- a cell release assay or animal model e.g ., mouse model
- Embodiments of the present invention include methods and related compositions for expressing and purifying the ArgRS protein fragments or other polypeptide-based agents of the invention.
- Such recombinant ArgRS polypeptides can be conveniently prepared using standard protocols as described for example in Sambrook, et al ., (1989, supra ), in particular Sections 16 and 17; Ausubel et al ., (1994, supra ), in particular Chapters 10 and 16; and Coligan et al., Current Protocols in Protein Science (John Wiley & Sons, Inc. 1995-1997 ), in particular Chapters 1, 5 and 6.
- ArgRS polypeptides may be prepared by a procedure including one or more of the steps of: (a) preparing a construct comprising a polynucleotide sequence that encodes a ArgRS polypeptide and that is operably linked to a regulatory element; (b) introducing the construct into a host cell; (c) culturing the host cell to express the ArgRS polypeptide; and (d) isolating the ArgRS polypeptide from the host cell.
- ArgRS polynucleotides are described elsewhere herein.
- a nucleotide sequence encoding the polypeptide, or a functional equivalent may be inserted into appropriate expression vector, i.e ., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence.
- appropriate expression vector i.e ., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence.
- Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding a polypeptide of interest and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described in Sambrook et al., Molecular Cloning, A Laboratory Manual (1989 ), and Ausubel et al., Current Protocols in Molecular Biology (1989 ).
- a variety of expression vector/host systems are known and may be utilized to contain and express polynucleotide sequences. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g ., baculovirus); plant cell systems transformed with virus expression vectors (e.g ., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g ., Ti or pBR322 plasmids); or animal cell systems, including mammalian cell and more specifically human cell systems.
- microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors
- yeast transformed with yeast expression vectors insect cell systems infected with virus expression vectors (e.g ., baculovirus)
- control elements or "regulatory sequences" present in an expression vector are those non-translated regions of the vector--enhancers, promoters, 5' and 3' untranslated regions--which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used.
- inducible promoters such as the hybrid lacZ promoter of the PBLUESCRIPT phagemid (Stratagene, La Jolla, Calif.) or PSPORT1 plasmid (Gibco BRL, Gaithersburg, Md.) and the like may be used.
- promoters from mammalian genes or from mammalian viruses are generally preferred. If it is necessary to generate a cell line that contains multiple copies of the sequence encoding a polypeptide, vectors based on SV40 or EBV may be advantageously used with an appropriate selectable marker.
- a number of expression vectors may be selected depending upon the use intended for the expressed polypeptide. For example, when large quantities are needed, vectors which direct high level expression of fusion proteins that are readily purified may be used.
- vectors include, but are not limited to, the multifunctional E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene), in which the sequence encoding the polypeptide of interest may be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of ⁇ -galactosidase so that a hybrid protein is produced; pIN vectors ( Van Heeke & Schuster, J. Biol. Chem.
- pGEX Vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
- GST glutathione S-transferase
- fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
- Proteins made in such systems may be designed to include heparin, thrombin, or factor XA protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.
- Certain embodiments may employ E. coli -based expression systems (see, e.g., Structural Genomics Consortium et al ., Nature Methods. 5:135-146, 2008 ). These and related embodiments may rely partially or totally on ligation-independent cloning (LIC) to produce a suitable expression vector.
- protein expression may be controlled by a T7 RNA polymerase (e.g ., pET vector series).
- T7 RNA polymerase e.g ., pET vector series
- These and related embodiments may utilize the expression host strain BL21(DE3), a ⁇ DE3 lysogen of BL21 that supports T7-mediated expression and is deficient in lon and ompT proteases for improved target protein stability.
- expression host strains carrying plasmids encoding tRNAs rarely used in E. coli such as ROSETTA TM (DE3) and Rosetta 2 (DE3) strains.
- Cell lysis and sample handling may also be improved using reagents sold under the trademarks BENZONASE® nuclease and BUGBUSTER® Protein Extraction Reagent.
- BENZONASE® nuclease e.g ., BUGBUSTER® Protein Extraction Reagent.
- auto-inducing media can improve the efficiency of many expression systems, including high-throughput expression systems.
- Media of this type e.g ., OVERNIGHT EXPRESSTM Autoinduction System gradually elicit protein expression through metabolic shift without the addition of artificial inducing agents such as IPTG.
- Particular embodiments employ hexahistidine tags (such as those sold under the trademark HIS•TAG® fusions), followed by immobilized metal affinity chromatography (IMAC) purification, or related techniques.
- clinical grade proteins can be isolated from E. coli inclusion bodies, without or without the use of affinity tags (see, e.g., Shimp et al., Proteins Expr Purif. 50:58-67, 2006 ).
- affinity tags see, e.g., Shimp et al., Proteins Expr Purif. 50:58-67, 2006 ).
- certain embodiments may employ a cold-shock induced E. coli high-yield production system, because over-expression of proteins in Escherichia coli at low temperature improves their solubility and stability ( see, e.g., Qing et al., Nature Biotechnology. 22:877-882, 2004 ).
- high-density bacterial fermentation systems For example, high cell density cultivation of Ralstonia eutropha allows protein production at cell densities of over 150 g/L, and the expression of recombinant proteins at titers exceeding 10 g/L.
- yeast Saccharomyces cerevisiae a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH may be used.
- constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH
- PGH palladium phosphate
- Pichia pandoris expression systems see, e.g., Li et al., Nature Biotechnology. 24, 210 - 215, 2006 ; and Hamilton et al., Science, 301:1244, 2003 ).
- yeast systems that are engineered to selectively glycosylate proteins, including yeast that have humanized N-glycosylation pathways, among others (see, e.g., Hamilton et al., Science. 313:1441-1443, 2006 ; Wildt et al., Nature Reviews Microbiol. 3:119-28, 2005 ; and Gerngross et al., Nature-Biotechnology. 22:1409 -1414, 2004 ; U.S. Patent Nos. 7,629,163 ; 7,326,681 ; and 7,029,872 ).
- recombinant yeast cultures can be grown in Fernbach Flasks or 15L, 50L, 100L, and 200L fermentors, among others.
- sequences encoding polypeptides may be driven by any of a number of promoters.
- viral promoters such as the 35S and 19S promoters of CaMV may be used alone or in combination with the omega leader sequence from TMV ( Takamatsu, EMBO J. 3:1671-1680 (1984 ); Broglie et al., Science 224:838-843 (1984 ); and Winter et al., Results Probl. Cell Differ. 17:85-105 (1991 )).
- constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. Such techniques are described in a number of generally available reviews (see, e.g ., Hobbs in McGraw Hill, Yearbook of Science and Technology, pp. 191-196 (1992 )).
- An insect system may also be used to express a polypeptide of interest.
- Autograph californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia ni cells.
- the sequences encoding the polypeptide may be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of the polypeptide-encoding sequence will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein.
- the recombinant viruses may then be used to infect, for example, S.
- frugiperda cells or Trichoplusia ni cells in which the polypeptide of interest may be expressed Engelhard et al., Pro. Natl. Acad. Sci. U.S.A. 91:3224-3227 (1994 )). Also included are baculovirus expression systems, including those that utilize SF9, SF21, and T. ni cells ( see, e.g., Murphy and Piwnica-Worms, Curr Proto Proteins Sci. Chapter 5:Unit5.4, 2001 ). Insect systems can provide post-translation modifications that are similar to mammalian systems.
- a number of viral-based expression systems are generally available.
- sequences encoding a polypeptide of interest may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain a viable virus which is capable of expressing the polypeptide in infected host cells ( Logan & Shenk, Proc. Natl. Acad. Sci. U.S.A. 81:3655-3659 (1984 )).
- transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.
- RSV Rous sarcoma virus
- Examples of useful mammalian host cell lines include monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells sub-cloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977 )); baby hamster kidney cells (BHK, ATCC CCL 10); mouse sertoli cells (TM4, Mather, Biol. Reprod.
- COS-7 monkey kidney CV1 line transformed by SV40
- human embryonic kidney line (293 or 293 cells sub-cloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977 )
- baby hamster kidney cells BHK, ATCC CCL 10
- mouse sertoli cells TM4, Mather, Biol. Reprod.
- monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TR1 cells ( Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982 )); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2).
- CHO Chinese hamster ovary
- DHFR-CHO cells Urlaub et al., PNAS USA 77:4216 (1980 )
- myeloma cell lines such as NSO and Sp2/0.
- CHO Chinese hamster ovary
- myeloma cell lines such as NSO and Sp2/0.
- Certain preferred mammalian cell expression systems include CHO and HEK293-cell based expression systems.
- Mammalian expression systems can utilize attached cell lines, for example, in T-flasks, roller bottles, or cell factories, or suspension cultures, for example, in 1L and 5L spinners, 5L, 14L, 40L, 100L and 200L stir tank bioreactors, or 20/50L and 100/200L WAVE bioreactors, among others known in the art.
- RNA polymerase typically utilize purified RNA polymerase, ribosomes, tRNA and ribonucleotides; these reagents may be produced by extraction from cells or from a cell-based expression system.
- Specific initiation signals may also be used to achieve more efficient translation of sequences encoding a polypeptide of interest. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding the polypeptide, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a portion thereof, is inserted, exogenous translational control signals including the ATG initiation codon should be provided. Furthermore, the initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used, such as those described in the literature ( Scharf. et al., Results Probl. Cell Differ. 20:125-162 (1994 )).
- a host cell strain may be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion.
- modifications of the polypeptide include, but are not limited to, post-translational modifications such as acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation.
- Post-translational processing which cleaves a "prepro" form of the protein may also be used to facilitate correct insertion, folding and/or function.
- Different host cells such as yeast, CHO, HeLa, MDCK, HEK293, and W138, in addition to bacterial cells, which have or even lack specific cellular machinery and characteristic mechanisms for such post-translational activities, may be chosen to ensure the correct modification and processing of the foreign protein.
- cell lines which stably express a polynucleotide of interest may be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for about 1-2 days in an enriched media before they are switched to selective media. The purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type. Transient production, such as by transient transfection or infection, can also be employed. Exemplary mammalian expression systems that are suitable for transient production include HEK293 and CHO-based systems.
- selection systems may be used to recover transformed or transduced cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase ( Wigler et al., Cell 11:223-232 (1977 )) and adenine phosphoribosyltransferase ( Lowy et al., Cell 22:817-823 (1990 )) genes which can be employed in tk- or aprt- cells, respectively. Also, antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dhfr which confers resistance to methotrexate ( Wigler et al., Pro. Natl. Acad. Sci. U.S.A.
- npt which confers resistance to the aminoglycosides, neomycin and G-418 ( Colbere-Garapin et al., J. Mol. Biol. 150:1-14 (1981 )); and als or pat, which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murry, supra). Additional selectable genes have been described, for example, trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine ( Hartman & Mulligan, Pro. Natl. Acad. Sci. U.S.A.
- GFP green fluorescent protein
- RFP red fluorescent protein
- YFP green fluorescent protein
- anthocyanins e.g., ⁇ -glucuronidase and its substrate GUS
- luciferase and its substrate luciferin being widely used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system (see, e.g., Rhodes et al., Methods Mol. Biol. 55:121-131 (1995 )).
- Embodiments of the present invention also include high-throughput protein production systems, or micro-production systems. Certain aspects may utilize, for example, hexa-histidine fusion tags for protein expression and purification on metal chelate-modified slide surfaces or MagneHis Ni-Particles ( see, e.g., Kwon et al., BMC Biotechnol. 9:72, 2009 ; and Lin et al., Methods Mol Biol. 498:129-41, 2009 )). Also included are high-throughput cell-free protein expression systems ( see, e.g., Sitaraman et al., Methods Mol Biol. 498:229-44, 2009 ). These and related embodiments can be used, for example, to generate microarrays of ArgRS protein fragment(s), which can then be used for screening libraries to identify agents that interact with the ArgRS protein fragment(s).
- Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides include oligolabeling, nick translation, end-labeling or PCR amplification using a labeled nucleotide.
- the sequences, or any portions thereof may be cloned into a vector for the production of an mRNA probe.
- Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides.
- reporter molecules or labels include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
- Host cells transformed with a polynucleotide sequence of interest may be cultured under conditions suitable for the expression and recovery of the protein from cell culture.
- Certain specific embodiments utilize serum free cell expression systems. Examples include HEK293 cells and CHO cells that can be grown on serum free medium ( see, e.g., Rosser et al., Protein Expr. Purif. 40:237-43, 2005 ; and U.S. Patent number 6,210,922 ).
- the protein produced by a recombinant cell may be secreted or contained intracellularly depending on the sequence and/or the vector used.
- expression vectors containing polynucleotides of the invention may be designed to contain signal sequences which direct secretion of the encoded polypeptide through a prokaryotic or eukaryotic cell membrane.
- Other recombinant constructions may be used to join sequences encoding a polypeptide of interest to nucleotide sequence encoding a polypeptide domain which will facilitate purification and / or detection of soluble proteins.
- cleavable and non-cleavable affinity purification and epitope tags such as avidin, FLAG tags, poly-histidine tags ( e.g ., 6xHis), cMyc tags, V5-tags, glutathione S-transferase (GST) tags, and others.
- the protein produced by a recombinant cell can be purified and characterized according to a variety of techniques known in the art.
- Exemplary systems for performing protein purification and analyzing protein purity include fast protein liquid chromatography (FPLC) (e.g ., AKTA and Bio-Rad FPLC systems), highpressure liquid chromatography (HPLC) (e.g ., Beckman and Waters HPLC).
- FPLC fast protein liquid chromatography
- HPLC highpressure liquid chromatography
- Exemplary chemistries for purification include ion exchange chromatography (e.g ., Q, S), size exclusion chromatography, salt gradients, affinity purification (e.g ., Ni, Co, FLAG, maltose, glutathione, protein A/G), gel filtration, reverse-phase, ceramic HYPERD® ion exchange chromatography, and hydrophobic interaction columns (HIC), among others known in the art. Also included are analytical methods such as SDS-PAGE (e.g ., coomassie, silver stain), immunoblot, Bradford, and ELISA, which may be utilized during any step of the production or purification process, typically to measure the purity of the protein composition.
- concentrated solutions of ArgRS polypeptides may comprise proteins at a concentration of about 5 mg/mL; or about 8 mg/mL; or about 10 mg/mL; about 15 mg/mL; or about 20 mg/mL.
- compositions may be substantially monodisperse, meaning that the ArgRS polypeptide compositions exist primarily (i.e. at least about 90%, or greater) in one apparent molecular weight form when assessed for example, by size exclusion chromatography, dynamic light scattering, or analytical ultracentrifugation.
- compositions have a purity (on a protein basis) of at least about 90%, or in some aspects at least about 95% purity, or in some embodiments, at least 98% purity. Purity may be determined via any routine analytical method as known in the art.
- compositions have a high molecular weight aggregate content of less than about 10%, compared to the total amount of protein present, or in some embodiments such compositions have a high molecular weight aggregate content of less than about 5%, or in some aspects such compositions have a high molecular weight aggregate content of less than about 3%, or in some embodiments a high molecular weight aggregate content of less than about 1%.
- High molecular weight aggregate content may be determined via a variety of analytical techniques including for example, by size exclusion chromatography, dynamic light scattering, or analytical ultracentrifugation.
- the ArgRS polypeptide compositions have an endotoxin content of less than about 10 EU / mg of ArgRS polypeptide, or less than about 5 EU / mg of ArgRS polypeptide, less than about 3 EU / mg of ArgRS polypeptide, or less than about 1 EU / mg of ArgRS polypeptide.
- concentration approaches contemplated herein include lyophilization, which is typically employed when the solution contains few soluble components other than the protein of interest. Lyophilization is often performed after HPLC run, and can remove most or all volatile components from the mixture. Also included are ultrafiltration techniques, which typically employ one or more selective permeable membranes to concentrate a protein solution. The membrane allows water and small molecules to pass through and retains the protein; the solution can be forced against the membrane by mechanical pump, gas pressure, or centrifugation, among other techniques.
- the reagents, ArgRS protein fragments, or related agents have a purity of at least about 90%, as measured according to routine techniques in the art.
- the ArgRS compositions of the present invention have a purity of at least about 95%.
- the ArgRS compositions of the present invention have a purity of at least about 97% or 98% or 99%.
- ArgRS protein fragments can be of lesser purity, and may have a purity of at least about 50%, 60%, 70%, or 80%. Purity can be measured overall or in relation to selected components, such as other proteins, e.g., purity on a protein basis.
- Purified ArgRS protein fragments can also be characterized according to their biological characteristics. Examples include binding affinity or binding kinetics to a selected ligand (e.g., a cellular binding partner of the ArgRS protein fragment such as a cell-surface receptor or an extracellular domain thereof), and the presence or levels of one or more canonical or non-canonical biological activity, as described herein. Binding affinity and binding kinetics can be measured according to a variety of techniques known in the art, such as Biacore® and related technologies that utilize surface plasmon resonance (SPR), an optical phenomenon that enables detection of unlabeled interactants in real time. SPR-based biosensors can be used in determination of active concentration, screening and characterization in terms of both affinity and kinetics.
- a selected ligand e.g., a cellular binding partner of the ArgRS protein fragment such as a cell-surface receptor or an extracellular domain thereof
- Binding affinity and binding kinetics can be measured according to a variety of techniques known in the art,
- the presence or levels of one or more canonical or non-canonical biological activities can be measured according to cell-based assays, including those that utilize a cellular binding partner (e.g ., cell-surface receptor) of a selected ArgRS protein fragment, which is functionally coupled to a readout or indicator, such as a fluorescent or luminescent indicator of a non-canonical biological activity, as described herein.
- a cellular binding partner e.g ., cell-surface receptor
- a readout or indicator such as a fluorescent or luminescent indicator of a non-canonical biological activity, as described herein.
- the ArgRS polypeptide compositions are about substantially endotoxin free, including, for example, about 95% endotoxin free, preferably about 99% endotoxin free, and more preferably about 99.99% endotoxin free.
- the presence of endotoxins can be detected according to routine techniques in the art, as described herein.
- the ArgRS compositions are made from a eukaryotic cell such as a mammalian or human cell in substantially serum free media.
- the ArgRS polypeptide compositions comprise less than about 10% wt/wt high molecular weight aggregates, or less than about 5% wt/wt high molecular weight aggregates, or less than about 2% wt/wt high molecular weight aggregates, or less than about or less than about 1% wt/wt high molecular weight aggregates.
- Protein-based analytical assays and methods which can be used to assess, for example, protein purity, size, solubility, and degree of aggregation, among other characteristics.
- Protein purity can be assessed a number of ways. For instance, purity can be assessed based on primary structure, higher order structure, size, charge, hydrophobicity, and glycosylation.
- methods for assessing primary structure include N- and C-terminal sequencing and peptide-mapping ( see, e.g., Allen et al., Biologicals. 24:255-275, 1996 )).
- methods for assessing higher order structure include circular dichroism ( see, e.g., Kelly et al., Biochim Biophys Acta.
- Hydrophobicity can be assessed, for example, by reverse-phase HPLC and hydrophobic interaction chromatography HPLC. Glycosylation can affect pharmacokinetics ( e.g ., clearance), conformation or stability, receptor binding, and protein function, and can be assessed, for example, by mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy.
- certain embodiments include the use of SEC-HPLC to assess protein characteristics such as purity, size (e.g., size homogeneity) or degree of aggregation, and/or to purify proteins, among other uses.
- SEC also including gel-filtration chromatography (GFC) and gel-permeation chromatography (GPC) refers to a chromatographic method in which molecules in solution are separated in a porous material based on their size, or more specifically their hydrodynamic volume, diffusion coefficient, and/or surface properties. The process is generally used to separate biological molecules, and to determine molecular weights and molecular weight distributions of polymers.
- a biological or protein sample (such as a protein extract produced according to the protein expression methods provided herein and known in the art) is loaded into a selected size-exclusion column with a defined stationary phase (the porous material), preferably a phase that does not interact with the proteins in the sample.
- the stationary phase is composed of inert particles packed into a dense three-dimensional matrix within a glass or steel column.
- the mobile phase can be pure water, an aqueous buffer, an organic solvent, or a mixture thereof.
- the stationary-phase particles typically have small pores and/or channels which only allow molecules below a certain size to enter.
- Protein purity for clinical applications is also discussed, for example, by Anicetti et al. (Trends in Biotechnology. 7:342-349, 1989 ). More recent techniques for analyzing protein purity include, without limitation, the LabChip GXII, an automated platform for rapid analysis of proteins and nucleic acids, which provides high throughput analysis of titer, sizing, and purity analysis of proteins.
- clinical grade proteins such as protein fragments and antibodies can be obtained by utilizing a combination of chromatographic materials in at least two orthogonal steps, among other methods ( see, e.g., Therapeutic Proteins: Methods and Protocols. Vol. 308, Eds., Smales and James, Humana Press Inc., 2005 ).
- protein agents e.g ., ArgRS protein fragments, antibodies, binding agents
- other agents e.g ., antisense, RNAi, small molecules
- protein agents e.g ., ArgRS protein fragments, antibodies, binding agents
- other agents e.g ., antisense, RNAi, small molecules
- Protein solubility assays are also included. Such assays can be utilized, for example, to determine optimal growth and purification conditions for recombinant production, to optimize the choice of buffer(s), and to optimize the choice of ArgRS protein fragments or variants thereof. Solubility or aggregation can be evaluated according to a variety of parameters, including temperature, pH, salts, and the presence or absence of other additives. Examples of solubility screening assays include, without limitation, microplate-based methods of measuring protein solubility using turbidity or other measure as an end point, high-throughput assays for analysis of the solubility of purified recombinant proteins ( see, e.g., Stenvall et al., Biochim Biophys Acta.
- ArgRS protein fragments with increased solubility can be identified or selected for according to routine techniques in the art, including simple in vivo assays for protein solubility (see, e.g., Maxwell et al., Protein Sci. 8:1908-11, 1999 ).
- Protein solubility and aggregation can also be measured by dynamic light scattering techniques.
- Aggregation is a general term that encompasses several types of interactions or characteristics, including soluble/insoluble, covalent/noncovalent, reversible/irreversible, and native/denatured interactions and characteristics.
- the presence of aggregates is typically considered undesirable because of the concern that aggregates may cause an immunogenic reaction (e.g ., small aggregates), or may cause adverse events on administration ( e.g ., particulates).
- Dynamic light scattering refers to a technique that can be used to determine the size distribution profile of small particles in suspension or polymers such as proteins in solution.
- This technique also referred to as photon correlation spectroscopy (PCS) or quasi-elastic light scattering (QELS), uses scattered light to measure the rate of diffusion of the protein particles. Fluctuations of the scattering intensity can be observed due to the Brownian motion of the molecules and particles in solution.
- This motion data can be conventionally processed to derive a size distribution for the sample, wherein the size is given by the Stokes radius or hydrodynamic radius of the protein particle. The hydrodynamic size depends on both mass and shape (conformation). Dynamic scattering can detect the presence of very small amounts of aggregated protein ( ⁇ 0.01% by weight), even in samples that contain a large range of masses.
- certain embodiments include the use of dynamic light scattering to analyze the solubility and/or presence of aggregates in a sample that contains an ArgRS protein fragment, antibody, or other agent of the invention.
- ArgRS agents such as ArgRS protein fragments, ArgRS polynucleotides, and antibodies and other binding agents described herein can be used in diagnostic assays and diagnostic compositions. Included are biochemical, histological, and cell-based methods and compositions, among others.
- ArgRS polypeptides include the detection of the ArgRS polynucleotide sequence(s) or corresponding ArgRS polypeptide sequence(s) or portions thereof of one or more newly identified ArgRS protein fragments, also referred to as ArgRS polypeptides.
- certain aspects include detection of the ArgRS polynucleotide sequence(s) or corresponding polypeptide sequence(s) or portions thereof of one or more newly identified ArgRS splice variants, and/or one or more splice junctions of those splice variants.
- the polynucleotide or corresponding polypeptide sequence(s) of at least one of the splice junctions is unique to that particular ArgRS splice variant.
- ArgRS protein fragments including splice variants, proteolytic fragments, and others.
- the presence or levels of one or more newly identified ArgRS protein fragments associate or correlate with one or more cellular types or cellular states.
- the presence or levels of an ArgRS polypeptide or polynucleotide can be used to distinguish between different cellular types or different cellular states.
- the presence or levels of ArgRS protein fragments or their related polynucleotides can be detected according to polynucleotide and/or polypeptide-based diagnostic techniques, as described herein and known in the art.
- Certain aspects can employ the ArgRS protein fragments, antibody, or ArgRS polynucleotides as part of a companion diagnostic method, typically to assess whether a subject or population subjects will respond favorably to a specific medical treatment.
- a given ArgRS therapeutic agent e.g ., protein fragment, antisense, RNAi, antibody, binding agent
- biomarkers include serum/tissue markers as well as markers that can be identified by medical imaging techniques.
- a naturally-occurring ArgRS protein fragment may itself provide a serum and/or tissue biomarker that can be utilized to measure drug outcome or assess the desirability of drug use in a specific subject or a specific population of subjects.
- the identification of an ArgRS polypeptide or polynucleotide reference sequence may include characterizing the differential expression of that sequence, whether in a selected subject, selected tissue, or otherwise, as described herein and known in the art.
- Certain of the methods provided herein rely on the differential expression of an ArgRS polypeptide or polynucleotide to characterize the condition or state of a cell, tissue, or subject, and to distinguish it from another cell, tissue, or subject.
- Non-limiting examples include methods of detecting the presence or levels of an ArgRS polypeptide or polynucleotide in a biological sample to distinguish between cells or tissues of different species, cells of different tissues or organs, cellular developmental states such as neonatal and adult, cellular differentiation states, conditions such as healthy, diseased and treated, intracellular and extracellular fractions, in addition to primary cell cultures and other cell cultures, such as immortalized cell cultures.
- Differential expression includes a statistically significant difference in one or more gene expression levels of an ArgRS polynucleotide or polypeptide reference sequence compared to the expression levels of the same sequence in an appropriate control.
- the statistically significant difference may relate to either an increase or a decrease in expression levels, as measured by RNA levels, protein levels, protein function, or any other relevant measure of gene expression such as those described herein.
- Also included is a comparison between an ArgRS polynucleotide or polypeptide of the invention and a full-length or wild-type cytosolic or mitochondrial ArgRS sequence, typically of the same or corresponding type.
- Differential expression can be detected by a variety of techniques in the art and described herein, including polynucleotide and polypeptide based techniques, such as real-time PCR, subtractive hybridization, polynucleotide and polypeptide arrays, and others.
- a result is typically referred to as statistically significant if it is unlikely to have occurred by chance.
- the significance level of a test or result relates traditionally to a frequentist statistical hypothesis testing concept.
- statistical significance may be defined as the probability of making a decision to reject the null hypothesis when the null hypothesis is actually true (a decision known as a Type I error, or "false positive determination"). This decision is often made using the p-value: if the p-value is less than the significance level, then the null hypothesis is rejected. The smaller the p-value, the more significant the result.
- Bayes factors may also be utilized to determine statistical significance ( see, e.g., Goodman S., Ann Intern Med 130:1005-13, 1999 ).
- the significance level of a test or result may reflect an analysis in which the probability of making a decision to reject the null hypothesis when the null hypothesis is actually true is no more than the stated probability. This type of analysis allows for those applications in which the probability of deciding to reject may be much smaller than the significance level for some sets of assumptions encompassed within the null hypothesis.
- statistically significant differential expression may include situations wherein the expression level of a given ArgRS sequence provides at least about a 1.2X, 1.3X, 1.4X, 1.5X, 1.6X, 1.7X, 1.8X, 1.9X, 2.0X, 2.2X, 2.4X, 2.6X, 2,8X, 3.0X, 4.0X, 5.0X, 6.0X, 7.0X, 8.0X, 9.0X, 10.0X, 15.0X, 20.0X, 50.0X, 100.0X, or greater difference in expression ( i.e., differential expression that may be higher or lower expression) in a suspected biological sample as compared to an appropriate control, including all integers and decimal points in between ( e.g., 1.24X, 1.25X, 2.1X, 2.5X, 60.0X, 75.0X, etc.).
- statistically significant differential expression may include situations wherein the expression level of a given ArgRS sequence provides at least about 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000 percent (%) or greater difference in expression (i.e ., differential expression that may be higher or lower) in a suspected biological sample as compared to an appropriate control, including all integers and decimal points in between.
- differential expression may also be determined by performing Z-testing, i.e., calculating an absolute Z score, as described herein and known in the art ( see Example 1).
- Z-testing is typically utilized to identify significant differences between a sample mean and a population mean. For example, as compared to a standard normal table (e.g., a control tissue), at a 95% confidence interval ( i.e., at the 5% significance level), a Z-score with an absolute value greater than 1.96 indicates non-randomness. For a 99% confidence interval, if the absolute Z is greater than 2.58, it means that p ⁇ .01, and the difference is even more significant-the null hypothesis can be rejected with greater confidence.
- an absolute Z-score of 1.96, 2, 2.58, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more, including all decimal points in between may provide a strong measure of statistical significance.
- an absolute Z-score of greater than 6 may provide exceptionally high statistical significance.
- Substantial similarly relates generally to the lack of a statistically significant difference in the expression levels between the biological sample and the reference control.
- substantially similar expression levels may include situations wherein the expression level of a given SSCIGS provides less than about a .05X, 0.1X, 0.2X, 0.3X, 0.4X, 0.5X, 0.6X, 0.7X, 0.8X, 0.9X, 1.0X, 1.1X, 1.2X, 1.3X, or 1.4X difference in expression (i.e., differential expression that may be higher or lower expression) in a suspected biological sample as compared to a reference sample, including all decimal points in between ( e.g., .15X, 0.25X, 0.35X, etc.).
- differential expression may include situations wherein the expression level of a given ArgRS sequence provides less than about 0.25. 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50 percent (%) difference in expression (i.e., differential expression that may be higher or lower) in a suspected biological sample as compared to a reference sample, including all decimal points in between.
- differential expression may also be determined by the mean expression value summarized by Affymetrix Microarray Suite 5 software (Affymetrix, Santa Clara, CA), or other similar software, typically with a scaled mean expression value of 1000.
- Embodiments of the present invention include methods of detecting the presence or levels of an ArgRS polynucleotide or polypeptide reference sequence or a portion thereof to distinguish between cells or tissues or other biological sample of a different organism or species, wherein the presence or levels of that sequence associates with a selected organism or species.
- General examples include methods of distinguishing between humans and any combination of bacteria, fungi, plants, and other non-human animals. Included within animals are methods of distinguishing between humans and any combination of vertebrates and invertebrates, including vertebrates such as fish, amphibians, reptiles, birds, and non-human mammals, and invertebrates such as insects, mollusks, crustaceans, and corals.
- non-human mammals are methods of distinguishing between humans and any combination of non-human mammals from the Order Afrosoricida, Macroscelidea, Tubulidentata, Hyracoidea, Proboscidea, Sirenia, Cingulata, Pilosa, Scandentia, Dermoptera, Primates, Rodentia, Lagomorpha, Erinaceomorpha, Soricomorpha, Chiroptera, Pholidota, Cetacea, Carnivora, Perissodactyla, or Artiodactyla.
- Primate Order are monkeys, apes, gorillas, and chimpanzees, among others known in the art.
- the presence or levels of an ArgRS polynucleotide or polypeptide reference sequence or variant, as described herein, may be used to identify the source of a given biological sample, such as a cell, tissue, or organ, by distinguishing between any combination of these organisms, or by distinguishing between humans and any one or more of these organisms, such as a panel of organisms.
- the source of a given biological sample may also be determined by comparing the presence or levels of an ArgRS sequence or a portion thereof to a pre-determined value.
- Embodiments of the present invention include methods of detecting the presence or levels of an ArgRS polynucleotide or polypeptide reference sequence or a portion thereof to distinguish between cells or other biological samples that originate from different tissues or organs.
- Non-limiting examples include methods of distinguishing between a cell or other biological sample that originates from any combination of skin (e.g., dermis, epidermis, subcutaneous layer), hair follicles, nervous system ( e.g., brain, spinal cord, peripheral nerves), auditory system or balance organs ( e.g., inner ear, middle ear, outer ear), respiratory system ( e.g., nose, trachea, lungs), gastroesophogeal tissues, the gastrointestinal system ( e.g., mouth, esophagus, stomach, small intestines, large intestines, rectum), vascular system ( e.g., heart, blood vessels and arteries), liver, gallbladder, lymphatic/immune system (
- Embodiments of the present invention include methods of detecting the presence or levels of an ArgRS polynucleotide or polypeptide reference sequence or a portion thereof to distinguish between or characterize the developmental or differentiation state of the cell. Also included are methods of differentiating between germ cells, stem cells, and somatic cells. Examples of developmental states include neonatal and adult. Examples of cellular differentiation states include all of the discreet and identifiable stages between a totipotent cell, a pluripotent cell, a multipotent progenitor stem cell and a mature, fully differentiated cell.
- a totipotent cell has total potential, typically arises during sexual and asexual reproduction, and includes and spores and zygotes, though in certain instances cells can dedifferentiate and regain totipotency.
- a pluripotent cell includes a stem cell that has the potential to differentiate into any of the three germ layers, including the endoderm (interior stomach lining, gastrointestinal tract, the lungs), the mesoderm (muscle, bone, blood, urogenital), and the ectoderm (epidermal tissues and nervous system).
- Multipotent progenitor cells are typically capable of differentiating into a limited number of tissue types.
- multipotent cells include, without limitation, hematopoietic stem cells (adult stem cells) from the bone marrow that give rise to immune cells such as red blood cells, white blood cells, and platelets, mesenchymal stem cells (adult stem cells) from the bone marrow that give rise to stromal cells, fat cells, and various types of bone cells, epithelial stem cells (progenitor cells) that give rise to the various types of skin cells, and muscle satellite cells (progenitor cells) that contribute to differentiated muscle tissue.
- hematopoietic stem cells adult stem cells
- immune cells such as red blood cells, white blood cells, and platelets
- mesenchymal stem cells adult stem cells
- progenitor cells epithelial stem cells
- muscle satellite cells progenitor cells
- ArgRS polynucleotide or polypeptide sequence e.g ., splice junction of an ArgRS splice variant, ArgRS proteolytic fragment
- splice junction of an ArgRS splice variant e.g ., ArgRS proteolytic fragment
- Embodiments of the present invention include methods of detecting the presence or levels of an ArgRS polynucleotide or polypeptide reference sequence to characterize or diagnose the condition or a cell, tissue, organ, or subject, in which that condition may be characterized as healthy, diseased, at risk for being diseased, or treated.
- diagnostic or “diagnosed” includes identifying the presence or nature of a pathologic condition, characterizing the risk of developing such a condition, and/or measuring the change (or no change) of a pathologic condition in response to therapy. Diagnostic methods may differ in their sensitivity and specificity.
- the "sensitivity" of a diagnostic assay refers to the percentage of diseased cells, tissues or subjects which test positive (percent of "true positives”). Diseased cells, tissues or subjects not detected by the assay are typically referred to as “false negatives.” Cells, tissues or subjects that are not diseased and which test negative in the assay may be termed “true negatives.”
- the "specificity" of a diagnostic assay may be defined as one (1) minus the false positive rate, where the "false positive” rate is defined as the proportion of those samples or subjects without the disease and which test positive. While a particular diagnostic method may not provide a definitive diagnosis of a condition, it suffices if the method provides a positive indication that aids in diagnosis.
- the presence or risk of developing a pathologic condition can be diagnosed by comparing the presence or levels of one or more selected ArgRS polynucleotide or polypeptide reference sequences or portions thereof that correlate with the condition, whether by increased or decreased levels, as compared to a suitable control.
- a "suitable control” or “appropriate control” includes a value, level, feature, characteristic, or property determined in a cell or other biological sample of a tissue or organism, e.g ., a control or normal cell, tissue or organism, exhibiting, for example, normal traits, such as the absence of the condition.
- a "suitable control” or “appropriate control” is a predefined value, level, feature, characteristic, or property.
- Other suitable controls will be apparent to persons skilled in the art. Examples of diseases and conditions are described elsewhere herein.
- Embodiments of the present invention include ArgRS polynucleotide or nucleic acid-based detection techniques, which offer certain advantages due to sensitivity of detection.
- certain embodiments relate to the use or detection of ArgRS polynucleotides as part of a diagnostic method or assay.
- the presence and/or levels of ArgRS polynucleotides may be measured by any method known in the art, including hybridization assays such as Northern blot, quantitative or qualitative polymerase chain reaction (PCR), quantitative or qualitative reverse transcriptase PCR (RT-PCR), microarray, dot or slot blots, or in situ hybridization such as fluorescent in situ hybridization (FISH), among others. Certain of these methods are described in greater detail below.
- ArgRS polynucleotides such as DNA and RNA can be collected and/or generated from blood, biological fluids, tissues, organs, cell lines, or other relevant sample using techniques known in the art, such as those described in Beverly. (2002 Current Protocols in Molecular Biology, Greene Publ. Assoc. Inc. & John Wiley & Sons, Inc., NY, NY ( see, e.g., as described by Nelson et al. Proc Natl Acad Sci U S A, 99: 11890-11895, 2002 ) and elsewhere. Further, a variety of commercially available kits for constructing RNA are useful for making the RNA to be used in the present invention.
- RNA may be constructed from organs/tissues/cells procured from normal healthy subjects; however, this invention also contemplates construction of RNA from diseased subjects. Certain embodiments contemplate using any type of organ from any type of subject or animal.
- RNA may be procured from an individual (e.g., any animal, including mammals) with or without visible disease and from tissue samples, biological fluids ( e.g., whole blood) or the like.
- RNA stabilizing reagents are optionally used, such as PAX tubes, as described, for example, in Thach et al., J. Immunol. Methods. Dec 283(1-2):269-279, 2003 and Chai et al., J. Clin. Lab Anal. 19(5):182-188, 2005 .
- Complementary DNA (cDNA) libraries can be generated using techniques known in the art, such as those described in Ausubel et al.
- Certain embodiments may employ hybridization methods for detecting ArgRS polynucleotide sequences.
- Methods for conducting polynucleotide hybridization assays have been well developed in the art. Hybridization assay procedures and conditions will vary depending on the application and are selected in accordance with the general binding methods known including those referred to in: Maniatis et al. Molecular Cloning: A Laboratory Manual (2nd Ed. Cold Spring Harbor, N.Y., 1989 ); Berger and Kimmel Methods in Enzymology, Vol. 152, Guide to Molecular Cloning Techniques (Academic Press, Inc., San Diego, Calif., 1987 ); Young and Davis, PNAS. 80: 1194 (1983 ). Methods and apparatus for carrying out repeated and controlled hybridization reactions have been described in U.S. Patent Nos. 5,871,928 , 5,874,219 , 6,045,996 and 6,386,749 , 6,391,623 .
- Certain embodiments may employ nucleic acid amplification methods for detecting ArgRS polynucleotide sequences.
- amplification or “nucleic acid amplification” refers to the production of multiple copies of a target nucleic acid that contains at least a portion of the intended specific target nucleic acid sequence. The multiple copies may be referred to as amplicons or amplification products.
- the amplified target contains less than the complete target gene sequence (introns and exons) or an expressed target gene sequence (spliced transcript of exons and flanking untranslated sequences).
- specific amplicons may be produced by amplifying a portion of the target polynucleotide by using amplification primers that hybridize to, and initiate polymerization from, internal positions of the target polynucleotide.
- the amplified portion contains a detectable target sequence that may be detected using any of a variety of well-known methods.
- Selective amplification or “specific amplification,” as used herein, refers to the amplification of a target nucleic acid sequence according to the present invention wherein detectable amplification of the target sequence is substantially limited to amplification of target sequence contributed by a nucleic acid sample of interest that is being tested and is not contributed by target nucleic acid sequence contributed by some other sample source, e.g ., contamination present in reagents used during amplification reactions or in the environment in which amplification reactions are performed.
- some other sample source e.g ., contamination present in reagents used during amplification reactions or in the environment in which amplification reactions are performed.
- amplification conditions refers to conditions permitting nucleic acid amplification according to the present invention.
- Amplification conditions may, in some embodiments, be less stringent than "stringent hybridization conditions" as described herein. Oligonucleotides used in the amplification reactions of the present invention hybridize to their intended targets under amplification conditions, but may or may not hybridize under stringent hybridization conditions. On the other hand, detection probes of the present invention typically hybridize under stringent hybridization conditions. Acceptable conditions to carry out nucleic acid amplifications according to the present invention can be easily ascertained by someone having ordinary skill in the art depending on the particular method of amplification employed.
- RNA amplification uses multiple cycles of denaturation, annealing of primer pairs to opposite strands, and primer extension to exponentially increase copy numbers of the target sequence.
- RT-PCR reverse transcriptase (RT) is used to make a complementary DNA (cDNA) from mRNA, and the cDNA is then amplified by PCR to produce multiple copies of DNA.
- PCR refers to multiple amplification cycles that selectively amplify a target nucleic acid species. Included are quantitative PCR (qPCR), real-time PCR), reverse transcription PCR (RT-PCR) and quantitative reverse transcription PCR (qRT-PCR) is well described in the art.
- qPCR quantitative PCR
- RT-PCR reverse transcription PCR
- qRT-PCR quantitative reverse transcription polymerase chain reaction
- pPCR quantitative polymerase chain reaction
- qRT-PCR refers to quantitative reverse transcription polymerase chain reaction.
- qPCR and qRT-PCR may be used to amplify and simultaneously quantify a targeted cDNA molecule. It enables both detection and quantification of a specific sequence in a cDNA pool, such as a selected ArgRS gene or transcript.
- real-time PCR may use DNA-binding dye to bind to all double-stranded (ds) DNA in PCR, causing fluorescence of the dye.
- dsDNA dyes such as SYBR Green will bind to all dsDNA PCR products. Fluorescence is detected and measured in the real-time PCR thermocycler, and its geometric increase corresponding to exponential increase of the product is used to determine the threshold cycle ("Ct") in each reaction.
- Ct Score refers to the threshold cycle number, which is the cycle at which PCR amplification has surpassed a threshold level. If there is a higher quantity of mRNA for a particular gene in a sample, it will cross the threshold earlier than a lowly expressed gene since there is more starting RNA to amplify. Therefore, a low Ct score indicates high gene expression in a sample and a high Ct score is indicative of low gene expression.
- Certain embodiments may employ the ligase chain reaction ( Weiss, Science. 254: 1292, 1991 ), commonly referred to as LCR, which uses two sets of complementary DNA oligonucleotides that hybridize to adjacent regions of the target nucleic acid.
- the DNA oligonucleotides are covalently linked by a DNA ligase in repeated cycles of thermal denaturation, hybridization and ligation to produce a detectable double-stranded ligated oligonucleotide product.
- SDA strand displacement amplification
- tSDA thermophilic endonucleases and polymerases at higher temperatures in essentially the same method
- amplification methods include, for example: nucleic acid sequence based amplification ( U.S. Pat. No. 5,130,238 ), commonly referred to as NASBA; one that uses an RNA replicase to amplify the probe molecule itself ( Lizardi, P. et al., 1988, BioTechnol. 6: 1197-1202 ), commonly referred to as Q ⁇ replicase; a transcription based amplification method ( Kwoh, D. et al., 1989, Proc. Natl. Acad. Sci. USA 86:1173-1177 ); self-sustained sequence replication ( Guatelli, J. et al., 1990, Proc. Natl. Acad. Sci.
- Illustrative transcription-based amplification systems of the present invention include TMA, which employs an RNA polymerase to produce multiple RNA transcripts of a target region ( U.S. Pat. Nos. 5,480,784 and 5,399,491 ).
- TMA uses a "promoter-primer" that hybridizes to a target nucleic acid in the presence of a reverse transcriptase and an RNA polymerase to form a double-stranded promoter from which the RNA polymerase produces RNA transcripts. These transcripts can become templates for further rounds of TMA in the presence of a second primer capable of hybridizing to the RNA transcripts.
- TMA is an isothermal method that uses an RNase H activity to digest the RNA strand of an RNA:DNA hybrid, thereby making the DNA strand available for hybridization with a primer or promoter-primer.
- RNase H activity associated with the reverse transcriptase provided for amplification is used.
- one amplification primer is an oligonucleotide promoter-primer that comprises a promoter sequence which becomes functional when double-stranded, located 5' of a target-binding sequence, which is capable of hybridizing to a binding site of a target RNA at a location 3' to the sequence to be amplified.
- a promoter-primer may be referred to as a "T7-primer" when it is specific for T7 RNA polymerase recognition. Under certain circumstances, the 3' end of a promoter-primer, or a subpopulation of such promoter-primers, may be modified to block or reduce primer extension.
- reverse transcriptase From an unmodified promoter-primer, reverse transcriptase creates a cDNA copy of the target RNA, while RNase H activity degrades the target RNA.
- a second amplification primer then binds to the cDNA. This primer may be referred to as a "non-T7 primer" to distinguish it from a "T7-primer.”
- reverse transcriptase creates another DNA strand, resulting in a double-stranded DNA with a functional promoter at one end.
- the promoter sequence is capable of binding an RNA polymerase to begin transcription of the target sequence to which the promoter-primer is hybridized.
- RNA polymerase uses this promoter sequence to produce multiple RNA transcripts (i.e., amplicons), generally about 100 to 1,000 copies. Each newly-synthesized amplicon can anneal with the second amplification primer. Reverse transcriptase can then create a DNA copy, while the RNase H activity degrades the RNA of this RNA:DNA duplex. The promoter-primer can then bind to the newly synthesized DNA, allowing the reverse transcriptase to create a double-stranded DNA, from which the RNA polymerase produces multiple amplicons. Thus, a billion-fold isothermic amplification can be achieved using two amplification primers.
- RNA transcripts of the transcripts from a particular cDNA library may be evaluated using other techniques, including microarray analysis ( Han, M., et al., Nat Biotechnol, 19: 631-635, 2001 ; Bao, P., et al., Anal Chem, 74: 1792-1797, 2002 ; Schena et al., Proc. Natl. Acad. Sci. USA 93:10614-19, 1996 ; and Heller et al., Proc. Natl. Acad. Sci. USA 94:2150-55, 1997 ) and SAGE (serial analysis of gene expression). Like MPSS, SAGE is digital and can generate a large number of signature sequences.
- the term "microarray” includes a "nucleic acid microarray” having a substrate-bound plurality of nucleic acids, hybridization to each of the plurality of bound nucleic acids being separately detectable.
- the substrate can be solid or porous, planar or non-planar, unitary or distributed.
- Nucleic acid microarrays include all the devices so called in Schena (ed.), DNA Microarrays: A Practical Approach (Practical Approach Series), Oxford University Press (1999 ); Nature Genet. 21(1) (suppl.): 1-60 (1999 ); Schena (ed.), Microarray Biochip: Tools and Technology, Eaton Publishing Company/BioTechniques Books Division (2000 ).
- Nucleic acid microarrays may include a substrate-bound plurality of nucleic acids in which the plurality of nucleic acids are disposed on a plurality of beads, rather than on a unitary planar substrate, as described, for example, in Brenner et al., Proc. Natl. Acad. Sci. USA 97(4): 1665-1670 (2000 ). Examples of nucleic acid microarrays may be found in U.S. Pat. Nos.
- nucleic acid arrays that are commercially available from Affymetrix (Santa Clara, Calif.) under the brand name GENECHIP TM . Further exemplary methods of manufacturing and using arrays are provided in, for example, US. Pat. Nos. 7,028,629 ; 7,011,949 ; 7,011,945 ; 6,936,419 ; 6,927,032 ; 6,924,103 ; 6,921,642 ; and 6,818,394 .
- the present invention as related to arrays and microarrays also contemplates many uses for polymers attached to solid substrates. These uses include gene expression monitoring, profiling, library screening, genotyping and diagnostics. Gene expression monitoring and profiling methods and methods useful for gene expression monitoring and profiling are shown in U.S. Pat. Nos. 5,800,992 , 6,013,449 , 6,020,135 , 6,033,860 , 6,040,138 , 6,177,248 and 6,309,822 . Genotyping and uses therefore are shown in U.S. Ser. Nos. 10/442,021 , 10/013,598 (U.S. Application No. 2003/0036069 ), and U.S. Pat. Nos.
- oligonucleotides such as primers or probes, for amplification or detection, as described herein.
- Oligonucleotides of a defined sequence and chemical structure may be produced by techniques known to those of ordinary skill in the art, such as by chemical or biochemical synthesis, and by in vitro or in vivo expression from recombinant nucleic acid molecules, e.g ., bacterial or viral vectors.
- an oligonucleotide does not consist solely of wild-type chromosomal DNA or the in vivo transcription products thereof.
- Oligonucleotides or primers may be modified in any way, as long as a given modification is compatible with the desired function of a given oligonucleotide.
- a given modification is suitable or desired for any given oligonucleotide of the present invention.
- Relevant ArgRS oligonucleotides are described in greater detail elsewhere herein.
- oligonucleotides While the design and sequence of oligonucleotides depends on their function as described herein, several variables are generally taken into account. Among the most relevant are: length, melting temperature (Tm), specificity, complementarity with other oligonucleotides in the system, G/C content, polypyrimidine (T, C) or polypurine (A, G) stretches, and the 3'-end sequence. Controlling for these and other variables is a standard and well known aspect of oligonucleotide design, and various computer programs are readily available to screen large numbers of potential oligonucleotides for optimal ones.
- Tm melting temperature
- specificity specificity
- G/C content polypyrimidine
- A, G polypurine
- Certain embodiments therefore include methods for detecting a target ArgRS polynucleotide in a sample, the polynucleotide comprising the sequence of a reference ArgRS polynucleotide, as described herein, comprising a) hybridizing the sample with a probe comprising a sequence complementary to the target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybridization complex, and optionally, if present, the amount thereof.
- Specific embodiments relate to the detection of ArgRS splice variants, such as by detecting a unique splice junction of the splice variant, whether by hybridization, amplification, or other detection method.
- Embodiments of the present invention include a variety of ArgRS polypeptide-based detection techniques, including antibody-based detection techniques. Included in these embodiments are the use of ArgRS polypeptides to generate antibodies or other binders, which may then be used in diagnostic methods and compositions to detect or quantitate selected ArgRS polypeptides in a cell or other biological sample, typically from a subject.
- Certain embodiments may employ standard methodologies and detectors such as western blotting and immunoprecipitation, enzyme-linked immunosorbent assays (ELISA), flow cytometry, and immunofluorescence assays (IFA), which utilize an imaging device.
- ELISA enzyme-linked immunosorbent assays
- IFA immunofluorescence assays
- These well-known methods typically utilize one or more monoclonal or polyclonal antibodies as described herein that specifically bind to a selected ArgRS polypeptide of the invention, or a unique region of that ArgRS polypeptide, and generally do not bind significantly to other ArgRS polypeptides, such as a full-length ArgRS polypeptide.
- the unique region of the ArgRS polypeptide may represent a unique three-dimensional structure that is possessed by a newly identified protein fragment of an ArgRS.
- a “microarray” may also refer to a "peptide microarray” or “protein microarray” having a substrate-bound collection or plurality of polypeptides, the binding to each of the plurality of bound polypeptides being separately detectable.
- the peptide microarray may have a plurality of binders, including but not limited to monoclonal antibodies, polyclonal antibodies, phage display binders, yeast 2 hybrid binders, and aptamers, which can specifically detect the binding of the ArgRS polypeptides described herein.
- the array may be based on autoantibody detection of these ArgRS polypeptides, as described, for example, in Robinson et al., Nature Medicine 8(3):295-301 (2002 ).
- Examples of peptide arrays may be found in WO 02/31463 , WO 02/25288 , WO 01/94946 , WO 01/88162 , WO 01/68671 , WO 01/57259 , WO 00/61806 , WO 00/54046 , WO 00/47774 , WO 99/40434 , WO 99/39210 , and WO 97/42507 and U.S. Pat. Nos. 6,268,210 , 5,766,960 , and 5,143,854 .
- MS or other molecular weight-based methods for diagnostically detecting ArgRS polypeptide sequences.
- Mass spectrometry refers generally to an analytical technique for determining the elemental composition of a sample or molecule. MS may also be used for determining the chemical structures of molecules, such as peptides and other chemical compounds.
- the MS principle consists of ionizing chemical compounds to generate charged molecules or molecule fragments, and then measuring their mass-to-charge ratios.
- a sample is loaded onto the MS instrument, and undergoes vaporization, the components of the sample are ionized by one of a variety of methods ( e.g., by impacting them with an electron beam), which results in the formation of positively charged particles, the positive ions are then accelerated by a magnetic field, computations are performed on the mass-to-charge ratio ( m / z ) of the particles based on the details of motion of the ions as they transit through electromagnetic fields, and, detection of the ions, which in step prior were sorted according to m / z.
- An illustrative MS instruments has three modules: an ion source, which converts gas phase sample molecules into ions (or, in the case of electrospray ionization, move ions that exist in solution into the gas phase); a mass analyzer, which sorts the ions by their masses by applying electromagnetic fields; and a detector, which measures the value of an indicator quantity and thus provides data for calculating the abundances of each ion present.
- an ion source which converts gas phase sample molecules into ions (or, in the case of electrospray ionization, move ions that exist in solution into the gas phase)
- a mass analyzer which sorts the ions by their masses by applying electromagnetic fields
- a detector which measures the value of an indicator quantity and thus provides data for calculating the abundances of each ion present.
- the MS technique has both qualitative and quantitative uses, including identifying unknown compounds, determining the isotopic composition of elements in a molecule, and determining the structure of a compound by observing its fragmentation.
- Other uses include quantifying the amount of a compound in a sample or studying the fundamentals of gas phase ion chemistry (the chemistry of ions and neutrals in a vacuum). Included are gas chromatography-mass spectrometry (GC/MS or GC-MS), liquid chromatography mass spectrometry (LC/MS or LC-MS), and ion mobility spectrometry/mass spectrometry (IMS/MS or IMMS). Accordingly, MS techniques may be used according to any of the methods provided herein to measure the presence or levels of an ArgRS polypeptide of the invention in a biological sample, and to compare those levels to a control sample or a pre-determined value.
- Certain embodiments may employ cell-sorting or cell visualization or imaging devices/techniques to detect or quantitate the presence or levels of ArgRS polynucleotides or polypeptides.
- Examples include flow cytometry or FACS, immunofluorescence analysis (IFA), and in situ hybridization techniques, such as fluorescent in situ hybridization (FISH).
- Computer software products of the invention typically include computer readable medium having computer-executable instructions for performing the logic steps of the method of the invention.
- Suitable computer readable medium include floppy disk, CD-ROM/DVD/DVD-ROM, hard-disk drive, flash memory, ROM/RAM, magnetic tapes and etc.
- the computer executable instructions may be written in a suitable computer language or combination of several languages.
- Certain embodiments may employ various computer program products and software for a variety of purposes, such as probe design, management of data, analysis, and instrument operation. See, U.S. Pat. Nos. 5,593,839 , 5,795,716 , 5,733,729 , 5,974,164 , 6,066,454 , 6,090,555 , 6,185,561 , 6,188,783 , 6,223,127 , 6,229,911 and 6,308,170 .
- WGSA whole genome sampling assay
- Kennedy et al. Nat. Biotech. 21, 1233-1237 (2003 )
- Matsuzaki et al. Gen. Res. 14: 414-425, (2004 )
- Matsuzaki, et al. Nature Methods 1:109-111 (2004 ).
- Algorithms for use with mapping assays are described, for example, in Liu et al., Bioinformatics. 19: 2397-2403 (2003 ) and Di et al. Bioinformatics. 21:1958 (2005 ). Additional methods related to WGSA and arrays useful for WGSA and applications of WGSA are disclosed, for example, in U.S. Patent Application Nos.
- certain embodiments may include methods for providing genetic information over networks such as the Internet as shown, for example, in U.S. Application Nos. 10/197,621 , 10/063,559 (United States Publication Number 2002/0183936 ), 10/065,856 , 10/065,868 , 10/328,818 , 10/328,872 , 10/423,403 , and 60/482,389 .
- Embodiments of the present invention also include antisense oligonucleotides and RNAi agents that target the ArgRS polynucleotide sequences, and methods of use thereof to reduce expression of a selected ArgRS transcript and/or protein fragment.
- Certain embodiments relate to targeting one or more splice junctions (often unique) that generate a splice variant, ArgRS protein fragment of instant invention.
- methods of antisense or RNAi inhibition that target certain splice forms, either to encourage or discourage splicing of a selected protein fragment.
- the splice junctions that generate the ArgRS protein fragments are over-expressed with respect to particular tissues, and are unique to that splice variant.
- such splice variants are not the only source of cytosolic ArgRS activity in the targeted cell type.
- certain splice variants to be targeted may represent about 10% to 50% of the total copy number of the ArgRS RNA splice variants in a given cell or tissue, and preferably about 1-10% of the total copy number of the ArgRS RNA splice variants in a given cell or tissue.
- Splice variants that are about ⁇ 1% of the total copy number of the ArgRS RNA splice variants in a given cell or tissue may also be targeted.
- the antisense or RNAi agent does not target the full-length protein, because such full-length proteins are responsible for a key step in protein synthesis, and thereby avoids lethality that often results from wild-type ArgRS knockouts.
- Certain of the methods described herein can therefore by used to avoid undesired effects such as toxicities in both chronic and acute treatments, and to selectively modulate the non-canonical activities of the ArgRS protein fragment.
- certain embodiments may generically target ArgRS sequences, including full-length ArgRS sequences, such as to kill or substantially derange the cell physiology of a target cell or tissue.
- the ArgRS splice variant to be targeted possesses a non-canonical biological activity.
- the ArgRS splice variant has reduced or undetectable canonical ArgRS activity, and the antisense or RNAi-related method more specifically modulates its non-canonical activity.
- the antisense or RNAi-related agents can be combined with a targeted or local delivery approach to lessen systemic undesired effects to non-targeted cells or tissues.
- exemplary cells or tissues that could be targeted this way include cancer cells, and cells to tissues that lend themselves to localized targeting, such as tumors or epithelia via topical application.
- antisense oligomer or “antisense compound” or “antisense oligonucleotide” are used interchangeably and refer to a sequence of cyclic subunits, each bearing a base-pairing moiety, linked by intersubunit linkages that allow the base-pairing moieties to hybridize to a target sequence in a nucleic acid (typically an RNA) by Watson-Crick base pairing, to form a nucleic acid:oligomer heteroduplex within the target sequence, and typically thereby prevent translation of that RNA. Also included are methods of use thereof to modulate expression of a selected ArgRS transcript, such as a splice variant or proteolytic fragment, and/or its corresponding polypeptide.
- Antisense oligonucleotides may contain between about 8 and 40 subunits, typically about 8-25 subunits, and preferably about 12 to 25 subunits. In certain embodiments, oligonucleotides may have exact sequence complementarity to the target sequence or near complementarity, as defined below. In certain embodiments, the degree of complementarity between the target and antisense targeting sequence is sufficient to form a stable duplex.
- the region of complementarity of the antisense oligomers with the target RNA sequence may be as short as 8-11 bases, but is preferably 12-15 bases or more, e.g., 12-20 bases, or 12-25 bases, including all integers in between these ranges.
- An antisense oligomer of about 14-15 bases is generally long enough to have a unique complementary sequence in targeting the selected ArgRS gene. In certain embodiments, a minimum length of complementary bases may be required to achieve the requisite binding Tm, as discussed herein.
- antisense oligomers as long as 40 bases may be suitable, where at least a minimum number of bases, e.g., 10-12 bases, are complementary to the target sequence.
- facilitated or active uptake in cells is optimized at oligomer lengths less than about 30.
- an optimum balance of binding stability and uptake generally occurs at lengths of 18-25 bases.
- antisense oligomers e.g., PNAs, LNAs, 2'-OMe, MOE
- PNAs, LNAs, 2'-OMe, MOE antisense oligomers
- LNAs, 2'-OMe, MOE antisense oligomers
- oligomers consist of about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 bases, in which at least about 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 contiguous or non-contiguous bases are complementary to their ArgRS target sequence, or variants thereof.
- antisense oligomers may be 100% complementary to an ArgRS nucleic acid target sequence, or it may include mismatches, e.g ., to accommodate variants, as long as a heteroduplex formed between the oligomer and ArgRS nucleic acid target sequence is sufficiently stable to withstand the action of cellular nucleases and other modes of degradation which may occur in vivo.
- target sequence refers to a portion of the target RNA against which the oligonucleotide is directed, that is, the sequence to which the oligonucleotide will hybridize by Watson-Crick base pairing of a complementary sequence.
- the target sequence may be a contiguous region of an ArgRS mRNA (e.g., a unique splice junction of an ArgRS mRNA), or may be composed of non-contiguous regions of the mRNA.
- Oligomer backbones which are less susceptible to cleavage by nucleases are discussed below. Mismatches, if present, are less destabilizing toward the end regions of the hybrid duplex than in the middle. The number of mismatches allowed will depend on the length of the oligomer, the percentage of G:C base pairs in the duplex, and the position of the mismatch(es) in the duplex, according to well understood principles of duplex stability. Although such an antisense oligomer is not necessarily 100% complementary to the ArgRS nucleic acid target sequence, it is effective to stably and specifically bind to the target sequence, such that a biological activity of the nucleic acid target, e.g ., expression of ArgRS protein(s), is modulated.
- a biological activity of the nucleic acid target e.g ., expression of ArgRS protein(s)
- the stability of the duplex formed between an oligomer and a target sequence is a function of the binding Tm and the susceptibility of the duplex to cellular enzymatic cleavage.
- the Tm of an antisense oligonucleotide with respect to complementary-sequence RNA may be measured by conventional methods, such as those described by Hames et al., Nucleic Acid Hybridization, IRL Press, 1985, pp.107-108 or as described in Miyada C.G. and Wallace R.B., 1987, Oligonucleotide hybridization techniques, Methods Enzymol. Vol. 154 pp. 94-107 .
- antisense oligomer may have a binding Tm, with respect to a complementary-sequence RNA, of greater than body temperature and preferably greater than 50°C. Tm's in the range 60-80°C or greater are preferred.
- the Tm of an oligomer compound, with respect to a complementary-based RNA hybrid can be increased by increasing the ratio of C:G paired bases in the duplex, and/or by increasing the length (in base pairs) of the heteroduplex.
- Antisense oligomers can be designed to block or inhibit translation of mRNA or to inhibit natural pre-mRNA splice processing, or induce degradation of targeted mRNAs, and may be said to be "directed to" or "targeted against” a target sequence with which it hybridizes.
- the target sequence may include any coding or non-coding sequence of an ArgRS mRNA transcript, and may thus by within an exon or within an intron.
- the target sequence is relatively unique or exceptional among ArgRSs (e.g., a full-length ArgRS) and is selective for reducing expression of a selected ArgRS protein fragment, such as a proteolytic fragment or splice variant.
- the target site includes a 3' or 5' splice site of a pre-processed mRNA, or a branch point.
- the target sequence for a splice site may include an mRNA sequence having its 5' end 1 to about 25 to about 50 base pairs downstream of a splice acceptor junction or upstream of a splice donor junction in a preprocessed mRNA.
- a target sequence may include a splice junction of an alternatively splice ArgRS mRNA, such as a splice junction that does not occur in the full-length ArgRS, or is unique or exceptional to that transcript, in that it either does not occur or only seldom occurs in other ArgRS splice variants.
- An oligomer is more generally said to be "targeted against" a biologically relevant target, such as reference ArgRS polynucleotide, when it is targeted against the nucleic acid of the target in the manner described herein.
- oligonucleotide is typically complementary to a target sequence, such as a target DNA or RNA.
- a target sequence such as a target DNA or RNA.
- complementary and complementarity refer to polynucleotides (i.e., a sequence of nucleotides) related by the base-pairing rules. For example, the sequence “A-G-T,” is complementary to the sequence “T-C-A.” Complementarity may be “partial,” in which only some of the nucleic acids' bases are matched according to the base pairing rules. Or, there may be “complete” or “total” complementarity (100%) between the nucleic acids.
- the degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. While perfect complementarity is often desired, some embodiments can include one or more but preferably 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 mismatches with respect to the target sequence. Variations at any location within the oligomer are included. In certain embodiments, variations in sequence near the termini of an oligomer are generally preferable to variations in the interior, and if present are typically within about 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 nucleotides of the 5' and/or 3' terminus.
- targeting sequence refers to the sequence in an oligonucleotide that is complementary (meaning, in addition, substantially complementary) to the target sequence in the DNA or RNA target molecule.
- the entire sequence, or only a portion, of the antisense compound may be complementary to the target sequence.
- the targeting sequence is formed of contiguous bases, but may alternatively be formed of non-contiguous sequences that when placed together, e.g ., from opposite ends of the oligonucleotide, constitute sequence that spans the target sequence.
- Target and targeting sequences are described as “complementary” to one another when hybridization occurs in an antiparallel configuration.
- a targeting sequence may have “near” or “substantial” complementarity to the target sequence and still function for the purpose of the present invention, that is, it may still be functionally “complementary.”
- an oligonucleotide may have at most one mismatch with the target sequence out of 10 nucleotides, and preferably at most one mismatch out of 20.
- an oligonucleotide may have at least about 80%, 85%, 90% sequence homology, and preferably at least 95% sequence homology, with an ArgRS reference polynucleotide sequence described herein, or its complement.
- An oligonucleotide "specifically hybridizes" to a target polynucleotide if the oligomer hybridizes to a target (e.g., an ArgRS reference polynucleotide or its complement) under physiological conditions, with a Tm substantially greater than 45°C, preferably at least 50°C, and typically 60°C-80°C or higher.
- a target e.g., an ArgRS reference polynucleotide or its complement
- Tm substantially greater than 45°C, preferably at least 50°C, and typically 60°C-80°C or higher.
- the Tm is the temperature at which 50% of a target sequence hybridizes to a complementary polynucleotide.
- the terms specifically binds or specifically hybridizes refer generally to an oligonucleotide probe or polynucleotide sequence that not only binds to its intended target gene sequence in a sample under selected hybridization conditions, but does not bind significantly to other target sequences in the sample, and thereby discriminates between its intended target and all other targets in the target pool.
- a probe that specifically hybridizes to its intended target sequence may also detect concentration differences under the selected hybridization conditions, as described herein.
- nuclease-resistant oligomeric molecule refers to one whose backbone is substantially resistant to nuclease cleavage, in non-hybridized or hybridized form; by common extracellular and intracellular nucleases in the body; that is, the oligomer shows little or no nuclease cleavage under normal nuclease conditions in the body to which the oligomer is exposed.
- heteroduplex refers to a duplex between an oligonucleotide and the complementary portion of a target polynucleotide, such as a target DNA or RNA.
- a “nuclease-resistant heteroduplex” refers to a heteroduplex formed by the binding of an oligomer to its complementary target, such that the heteroduplex is substantially resistant to in vivo degradation by intracellular and extracellular nucleases, such as RNaseH, which are capable of cutting double-stranded RNA/RNA or RNA/DNA complexes.
- a “subunit" of an oligonucleotide refers to one nucleotide (or nucleotide analog) unit.
- the term may refer to the nucleotide unit with or without the attached intersubunit linkage, although, when referring to a "charged subunit", the charge typically resides within the intersubunit linkage (e.g., a phosphate or phosphorothioate linkage or a cationic linkage).
- oligonucleotide may be based on ribose or another pentose sugar or, in certain embodiments, alternate or modified groups.
- modified oligonucleotide backbones include, without limitation, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to
- PNAs peptide nucleic acids
- LNAs locked nucleic acids
- MOE 2'-methoxyethoxy oligonucleotides
- the purine or pyrimidine base pairing moiety is typically adenine, cytosine, guanine, uracil, thymine or inosine. Also included are bases such as pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2,4,6-trime115thoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines ( e.g., 5-methylcytidine), 5-alkyluridines ( e.g., ribothymidine), 5-halouridine ( e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g.
- 6-methyluridine 6-methyluridine
- propyne quesosine, 2-thiouridine, 4-thiouridine, wybutosine, wybutoxosine, 4-acetyltidine, 5-(carboxyhydroxymethyl)uridine, 5'-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluridine, ⁇ -D-galactosylqueosine, 1-methyladenosine, 1-methylinosine, 2,2-dimethylguanosine, 3-methylcytidine, 2-methyladenosine, 2-methylguanosine, N6-methyladenosine, 7-methylguanosine, 5-methoxyaminomethyl-2-thiouridine, 5-methylaminomethyluridine, 5-methylcarbonyhnethyluridine, 5-methyloxyuridine, 5-methyl-2-thiouridine, 2-methylthio-N6-isopentenyladenosine, ⁇ -D-mannosylqueosine
- modified bases in this aspect is meant nucleotide bases other than adenine (A), guanine (G), cytosine (C), thymine (T), and uracil (U), as illustrated above; such bases can be used at any position in the antisense molecule.
- Ts and Us are interchangeable. For instance, with other antisense chemistries such as 2'-O-methyl antisense oligonucleotides that are more RNA-like, the T bases may be shown as U.
- PNAs peptide nucleic acids
- PNAs are analogs of DNA in which the backbone is structurally homomorphous with a deoxyribose backbone, consisting of N-(2-aminoethyl) glycine units to which pyrimidine or purine bases are attached.
- PNAs containing natural pyrimidine and purine bases hybridize to complementary oligonucleotides obeying Watson-Crick base-pairing rules, and mimic DNA in terms of base pair recognition (Egholm, Buchardt et al. 1993).
- the backbone of PNAs is formed by peptide bonds rather than phosphodiester bonds, making them well-suited for antisense applications (see structure below).
- the backbone is uncharged, resulting in PNA/DNA or PNA/RNA duplexes that exhibit greater than normal thermal stability. PNAs are not recognized by nucleases or proteases.
- PNAs may be produced synthetically using any technique known in the art.
- PNA is a DNA analog in which a polyamide backbone replaces the traditional phosphate ribose ring of DNA.
- PNA is capable of sequence-specific binding in a helix form to DNA or RNA.
- Characteristics of PNA include a high binding affinity to complementary DNA or RNA, a destabilizing effect caused by single-base mismatch, resistance to nucleases and proteases, hybridization with DNA or RNA independent of salt concentration and triplex formation with homopurine DNA.
- PanageneTM has developed its proprietary Bts PNA monomers (Bts; benzothiazole-2-sulfonyl group) and proprietary oligomerisation process.
- the PNA oligomerisation using Bts PNA monomers is composed of repetitive cycles of deprotection, coupling and capping.
- Panagene's patents to this technology include US 6969766 , US 7211668 , US 7022851 , US 7125994 , US 7145006 and US 7179896 .
- Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082 ; 5,714,331 ; and 5,719,262 . Further teaching of PNA compounds can be found in Nielsen et al., Science, 1991, 254, 1497 .
- LNAs locked nucleic acid subunits
- the structures of LNAs are known in the art: for example, Wengel, et al., Chemical Communications (1998) 455 ; Tetrahedron (1998) 54, 3607 , and Accounts of Chem. Research (1999) 32, 301 ); Obika, et al., Tetrahedron Letters (1997) 38, 8735 ; ( 1998) 39, 5401 , and Bioorganic Medicinal Chemistry (2008)16, 9230 .
- Oligonucleotides may incorporate one or more LNAs; in some cases, the compounds may be entirely composed of LNAs.
- Methods for the synthesis of individual LNA nucleoside subunits and their incorporation into oligonucleotides are known in the art: U.S. Patents 7,572,582 ; 7,569,575 ; 7,084,125 ; 7,060,809 ; 7,053,207 ; 7,034,133 ; 6,794,499 ; and 6,670,461 .
- Typical intersubunit linkers include phosphodiester and phosphorothioate moieties; alternatively, non-phosphorous containing linkers may be employed.
- a preferred embodiment is an LNA containing compound where each LNA subunit is separated by a DNA subunit (i.e., a deoxyribose nucleotide). Further preferred compounds are composed of alternating LNA and DNA subunits where the intersubunit linker is phosphorothioate.
- oligonucleotides may comprise morpholino-based subunits bearing base-pairing moieties, joined by uncharged or substantially uncharged linkages.
- morpholino oligomer or "PMO” (phosphoramidate- or phosphorodiamidate morpholino oligomer) refer to an oligonucleotide analog composed of morpholino subunit structures, where (i) the structures are linked together by phosphorus-containing linkages, one to three atoms long, preferably two atoms long, and preferably uncharged or cationic, joining the morpholino nitrogen of one subunit to a 5' exocyclic carbon of an adjacent subunit, and (ii) each morpholino ring bears a purine or pyrimidine or an equivalent base-pairing moiety effective to bind, by base specific hydrogen bonding, to a base in a polynucleotide.
- the oxygen attached to phosphorus may be substituted with sulfur (thiophosphorodiamidate).
- the 5' oxygen may be substituted with amino or lower alkyl substituted amino.
- the pendant nitrogen attached to phosphorus may be unsubstituted, monosubstituted, or disubstituted with (optionally substituted) lower alkyl.
- the purine or pyrimidine base pairing moiety is typically adenine, cytosine, guanine, uracil, thymine or inosine.
- the morpholino subunits may also be linked by non-phosphorus-based intersubunit linkages, as described further below, where at least one linkage is modified with a pendant cationic group as described above.
- Other oligonucleotide analog linkages which are uncharged in their unmodified state but which could also bear a pendant amine substituent could be used.
- a 5'nitrogen atom on a morpholino ring could be employed in a sulfamide linkage or a urea linkage (where phosphorus is replaced with carbon or sulfur, respectively) and modified in a manner analogous to the 5'-nitrogen atom in structure (b3) above
- substantially uncharged morpholino oligomers such as a substantially uncharged phosphorodiamidate-linked morpholino oligomer.
- a substantially uncharged, phosphorus containing backbone in an oligonucleotide analog is one in which a majority of the subunit linkages, e.g ., between 50-100%, typically at least 60% to 100% or 75% or 80% of its linkages, are uncharged at physiological pH, and contain a single phosphorous atom.
- Examples of morpholino oligonucleotides having phosphorus-containing backbone linkages include phosphoroamidate and phosphorodiamidate-linked morpholino oligonucleotides.
- Certain embodiments may contain positively charged groups at preferably about 10%-50% of their backbone linkages.
- Properties of the morpholino-based subunits include, for example, the ability to be linked in a oligomeric form by stable, uncharged or positively charged backbone linkages, the ability to support a nucleotide base (e.g., adenine, cytosine, guanine, thymidine, uracil and hypoxanthine) such that the polymer formed can hybridize with a complementary-base target nucleic acid, including target RNA, Tm values above about 45°C in relatively short oligonucleotides ( e.g., 10-15 bases), the ability of the oligonucleotide to be actively or passively transported into mammalian cells, and the ability of the antisense oligonucleotide:RNA heteroduplex to resist RNase and RNaseH degradation, respectively.
- a nucleotide base e.g., adenine, cytosine, guanine, thymidine, uracil and hypoxanthin
- a substantially uncharged oligonucleotide may be modified to include charged linkages, e.g ., up to about 1 per every 2-5 uncharged linkages, such as about 4-5 per every 10 uncharged linkages.
- optimal improvement in antisense activity may be seen when about 25% of the backbone linkages are cationic.
- enhancement may be seen with a small number e.g ., 10-20% cationic linkages, or where the number of cationic linkages are in the range 50-80%, such as about 60%.
- the cationic backbone charges may be further enhanced by distributing the bulk of the charges close of the "center-region" backbone linkages of the antisense oligonucleotide, e.g., in a 20-mer oligonucleotide with 8 cationic backbone linkages, having at least 70% of these charged linkages localized in the 10 centermost linkages.
- Oligonucleotides that target one or more portions of an ArgRS polynucleotide reference sequence or its complement may be used in any of the therapeutic, diagnostic, or drug screening methods described herein and apparent to persons skilled in the art.
- RNA interference agents that target one or more mRNA transcripts of an arginyl aminoacyl-tRNA synthetase (ArgRS) reference polynucleotide, including fragments and splice variants thereof. Also included are methods of use thereof to modulate the levels of a selected ArgRS transcript, such as an ArgRS splice variant or endogenous proteolytic fragment.
- ArgRS arginyl aminoacyl-tRNA synthetase
- double-stranded means two separate nucleic acid strands comprising a region in which at least a portion of the strands are sufficiently complementary to hydrogen bond and form a duplex structure.
- duplex or “duplex structure” refers to the region of a double stranded molecule wherein the two separate strands are substantially complementary, and thus hybridize to each other.
- dsRNA refers to a ribonucleic acid molecule having a duplex structure comprising two complementary and anti-parallel nucleic acid strands ( i.e., the sense and antisense strands). Not all nucleotides of a dsRNA must exhibit Watson-Crick base pairs; the two RNA strands may be substantially complementary. The RNA strands may have the same or a different number of nucleotides.
- a dsRNA is or includes a region which is at least partially complementary to the target RNA. In certain embodiments, the dsRNA is fully complementary to the target RNA. It is not necessary that there be perfect complementarity between the dsRNA and the target, but the correspondence must be sufficient to enable the dsRNA, or a cleavage product thereof, to direct sequence specific silencing, such as by RNAi cleavage of the target RNA. Complementarity, or degree of homology with the target strand, is typically most critical in the antisense strand.
- some embodiments can include one or more but preferably 6, 5, 4, 3, 2, or fewer mismatches with respect to the target RNA.
- the mismatches are most tolerated in the terminal regions, and if present are preferably in a terminal region or regions, e.g ., within 6, 5, 4, or 3 nucleotides of the 5' and/or 3' terminus.
- the sense strand need only be substantially complementary with the antisense strand to maintain the overall double-strand character of the molecule.
- modified dsRNA refers to a dsRNA molecule that comprises at least one alteration that renders it more resistant to nucleases (e.g., protein kinase) than an identical dsRNA molecule that recognizes the same target RNA.
- Modified dsRNAs may include a single-stranded nucleotide overhang and/or at least one substituted nucleotide.
- nucleotide overhang refers to the unpaired nucleotide or nucleotides that protrude from the duplex structure when a 3'-end of one RNA strand extends beyond the 5'-end of the other complementary strand, or vice versa.
- Bount or “blunt end” means that there are no unpaired nucleotides at that end of the dsRNA, i.e ., no nucleotide overhang.
- a “blunt ended" dsRNA is a dsRNA that is double stranded over its entire length, i.e., no nucleotide overhang at either end of the molecule.
- terminal base pair refers to the last nucleotide base pair on one end of the duplex region of a double-stranded molecule.
- a dsRNA or other molecule is blunt ended (i.e., has no nucleotide overhangs)
- the last nucleotide base pairs at both ends of the molecule are terminal base pairs.
- the last nucleotide base pair(s) immediately adjacent the nucleotide overhang(s) is the terminal base pair at that end(s) of the molecule.
- the methods provided herein may utilize double-stranded ribonucleic acid (dsRNA) molecules as modulating agents, for reducing expression of an ArgRS transcript such as a selected fragment or splice variant.
- dsRNAs generally comprise two single strands. One strand of the dsRNA comprises a nucleotide sequence that is substantially identical to a portion of the target gene or target region (the "sense” strand), and the other strand (the “complementary” or “antisense” strand) comprises a sequence that is substantially complementary to a portion of the target region. The strands are sufficiently complementary to hybridize to form a duplex structure.
- the complementary RNA strand may be less than 30 nucleotides, less than 25 nucleotides in length, or even 19 to 24 nucleotides in length. In certain aspects, the complementary nucleotide sequence may be 20-23 nucleotides in length, or 22 nucleotides in length.
- RNA strands comprises a nucleotide overhang of 1 to 4 nucleotides in length.
- the dsRNA may further comprise at least one chemically modified nucleotide.
- a dsRNA comprising a single-stranded overhang of 1 to 4 nucleotides may comprise a molecule wherein the unpaired nucleotide of the single-stranded overhang that is directly adjacent to the terminal nucleotide pair contains a purine base.
- the last complementary nucleotide pairs on both ends of a dsRNA are a G-C pair, or, at least two of the last four terminal nucleotide pairs are G-C pairs.
- Micro-RNAs represent a large group of small RNAs produced naturally in organisms, some of which regulate the expression of target genes. Micro-RNAs are formed from an approximately 70 nucleotide single-stranded hairpin precursor transcript by Dicer. ( V. Ambros et al. Current Biology 13:807, 2003 ). Certain micro-RNAs may be transcribed as hairpin RNA precursors, which are then processed to their mature forms by Dicer enzyme.
- the first strand of the double-stranded oligonucleotide contains two more nucleoside residues than the second strand.
- the first strand and the second strand have the same number of nucleosides; however, the first and second strands may be offset such that the two terminal nucleosides on the first and second strands are not paired with a residue on the complimentary strand.
- the two nucleosides that are not paired are thymidine resides.
- shRNAs short hairpin RNAs
- miRNAs micro RNAs
- shRNAs short hairpin RNAs
- miRNAs micro RNAs
- a double-stranded structure of an shRNA is formed by a single self-complementary RNA strand, and RNA duplex formation may be initiated either inside or outside the cell.
- miRNAs are small non-coding RNAs of 20-22 nucleotides, typically excised from ⁇ 70 nucleotide foldback RNA precursor structures known as pre-miRNAs.
- the agent should include a region of sufficient homology to the target region, and be of sufficient length in terms of nucleotides, such that the siRNA agent, or a fragment thereof, can mediate down regulation of the target RNA.
- the term "ribonucleotide” or “nucleotide” can, in the case of a modified RNA or nucleotide surrogate, also refer to a modified nucleotide, or surrogate replacement moiety at one or more positions.
- an siRNA agent is or includes a region which is at least partially complementary to the target RNA, as described herein.
- an siRNA modulating agent may be modified or include nucleoside surrogates.
- Single stranded regions of an siRNA agent may be modified or include nucleoside surrogates, e.g ., the unpaired region or regions of a hairpin structure, e.g., a region which links two complementary regions, can have modifications or nucleoside surrogates. Modification to stabilize one or more 3'- or 5'-terminus of an siRNA agent, e.g ., against exonucleases, or to favor the antisense siRNA agent to enter into RISC are also useful.
- Modifications can include C3 (or C6, C7, C12) amino linkers, thiol linkers, carboxyl linkers, non-nucleotidic spacers (C3, C6, C9, C12, abasic, triethylene glycol, hexaethylene glycol), special biotin or fluorescein reagents that come as phosphoramidites and that have another DMT-protected hydroxyl group, allowing multiple couplings during RNA synthesis.
- siRNA agents may include, for example, molecules that are long enough to trigger the interferon response (which can be cleaved by Dicer ( Bernstein et al. 2001. Nature, 409:363-366 ) and enter a RISC (RNAi-induced silencing complex)), in addition to molecules which are sufficiently short that they do not trigger the interferon response (which molecules can also be cleaved by Dicer and/or enter a RISC), e.g ., molecules which are of a size which allows entry into a RISC, e.g ., molecules which resemble Dicer-cleavage products.
- RISC RNAi-induced silencing complex
- An siRNA modulating agent or a cleavage product thereof, can down regulate a target gene, e.g., by inducing RNAi with respect to a target RNA, preferably an ArgRS target such as a selected splice variant.
- Each strand of an siRNA agent can be equal to or less than 35, 30, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, or 15 nucleotides in length.
- the strand is preferably at least 19 nucleotides in length.
- each strand can be between 21 and 25 nucleotides in length.
- Preferred siRNA agents have a duplex region of 17, 18, 19, 29, 21, 22, 23, 24, or 25 nucleotide pairs, and one or more overhangs, preferably one or two 3' overhangs, of 2-3 nucleotides.
- an siRNA agent may have one or more of the following properties: it may, despite modifications, even to a very large number, or all of the nucleosides, have an antisense strand that can present bases (or modified bases) in the proper three dimensional framework so as to be able to form correct base pairing and form a duplex structure with a homologous target RNA which is sufficient to allow down regulation of the target, e.g., by cleavage of the target RNA; it may, despite modifications, even to a very large number, or all of the nucleosides, still have "RNA-like" properties, i.e., it may possess the overall structural, chemical and physical properties of an RNA molecule, even though not exclusively, or even partly, of ribonucleotide-based content.
- an siRNA agent can contain, e.g., a sense and/or an antisense strand in which all of the nucleotide sugars contain e.g., 2' fluoro in place of 2' hydroxyl.
- This deoxyribonucleotide-containing agent can still be expected to exhibit RNA-like properties.
- the electronegative fluorine prefers an axial orientation when attached to the C2' position of ribose. This spatial preference of fluorine can, in turn, force the sugars to adopt a C 3 '-endo pucker. This is the same puckering mode as observed in RNA molecules and gives rise to the RNA-characteristic A-family-type helix.
- fluorine is a good hydrogen bond acceptor, it can participate in the same hydrogen bonding interactions with water molecules that are known to stabilize RNA structures. Generally, it is preferred that a modified moiety at the 2' sugar position will be able to enter into H-bonding which is more characteristic of the OH moiety of a ribonucleotide than the H moiety of a deoxyribonucleotide.
- a "single strand RNAi agent” as used herein, is an RNAi agent which is made up of a single molecule. It may include a duplexed region, formed by intra-strand pairing, e.g., it may be, or include, a hairpin or pan-handle structure.
- Single strand RNAi modulating agents are preferably antisense with regard to the target molecule.
- a single strand RNAi agent should be sufficiently long that it can enter the RISC and participate in RISC mediated cleavage of a target mRNA.
- a single strand RNAi agent is at least 14, and more preferably at least 15, 20, 25, 29, 35, 40, or 50 nucleotides in length. It is preferably less than 200, 100, or 60 nucleotides in length.
- Hairpin RNAi modulating agents may have a duplex region equal to or at least 17, 18, 19, 29, 21, 22, 23, 24, or 25 nucleotide pairs.
- the duplex region may preferably be equal to or less than 200, 100, or 50, in length. Certain ranges for the duplex region are 15-30, 17 to 23, 19 to 23, and 19 to 21 nucleotides pairs in length.
- the hairpin may have a single strand overhang or terminal unpaired region, preferably the 3', and preferably of the antisense side of the hairpin. In certain embodiments, overhangs are 2-3 nucleotides in length.
- RNAi oligonucleotides such as chimeric oligonucleotides, or "chimeras,” which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound.
- oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
- Chimeric oligonucleotides may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleotides and/or oligonucleotide mimetics as described above. Such oligonucleotides have also been referred to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Pat. Nos.
- the chimeric oligonucleotide is RNA-DNA, DNA-RNA, RNA-DNA-RNA, DNA-RNA-DNA, or RNA-DNA-RNA-DNA, wherein the oligonucleotide is between 5 and 60 nucleotides in length.
- RNAi agents relate to an oligonucleotide comprising at least one ligand tethered to an altered or non-natural nucleobase.
- a large number of compounds can function as the altered base.
- the structure of the altered base is important to the extent that the altered base should not substantially prevent binding of the oligonucleotide to its target, e.g ., mRNA.
- the altered base is difluorotolyl, nitropyrrolyl, nitroimidazolyl, nitroindolyl, napthalenyl, anthrancenyl, pyridinyl, quinolinyl, pyrenyl, or the divalent radical of any one of the non-natural nucleobases described herein.
- the non-natural nucleobase is difluorotolyl, nitropyrrolyl, or nitroimidazolyl.
- the non-natural nucleobase is difluorotolyl.
- a wide variety of ligands are known in the art and are amenable to the present invention.
- the ligand can be a steroid, bile acid, lipid, folic acid, pyridoxal, B12, riboflavin, biotin, aromatic compound, polycyclic compound, crown ether, intercalator, cleaver molecule, protein-binding agent, or carbohydrate.
- the ligand is a steroid or aromatic compound.
- the ligand is cholesteryl.
- the RNAi agent is an oligonucleotide tethered to a ligand for the purposes of improving cellular targeting and uptake.
- a ligand for the purposes of improving cellular targeting and uptake.
- an RNAi agent may be tethered to an antibody, or antigen binding fragment thereof.
- an RNAi agent may be tethered to a specific ligand binding molecule, such as a polypeptide or polypeptide fragment that specifically binds a particular cell-surface receptor.
- the modulating agent comprises a non-natural nucleobase, as described herein.
- the ribose sugar moiety that naturally occurs in nucleosides is replaced with a hexose sugar.
- the hexose sugar is an allose, altrose, glucose, mannose, gulose, idose, galactose, talose, or a derivative thereof.
- the hexose is a D-hexose.
- the ribose sugar moiety that naturally occurs in nucleosides is replaced with a polycyclic heteroalkyl ring or cyclohexenyl group.
- the polycyclic heteroalkyl group is a bicyclic ring containing one oxygen atom in the ring.
- the polycyclic heteroalkyl group is a bicyclo[2.2.1]heptane, a bicyclo[3.2.1]octane, or a bicyclo[3.3.1]nonane.
- modified RNAi agents also include oligonucleotides containing modified backbones or non-natural internucleoside linkages, as described herein.
- the present invention further encompasses oligonucleotides employing ribozymes.
- Synthetic RNA molecules and derivatives thereof that catalyze highly specific endoribonuclease activities are known as ribozymes.
- the cleavage reactions are catalyzed by the RNA molecules themselves.
- the sites of self-catalyzed cleavage are located within highly conserved regions of RNA secondary structure ( Buzayan et al., Proc. Natl. Acad. Sci.
- RNA molecules Naturally occurring autocatalytic RNA molecules have been modified to generate ribozymes which can be targeted to a particular cellular or pathogenic RNA molecule with a high degree of specificity.
- ribozymes serve the same general purpose as antisense oligonucleotides (i.e., modulation of expression of a specific gene) and, like oligonucleotides, are nucleic acids possessing significant portions of single-strandedness.
- RNAi agents or antisense oligonucleotides for use with the methods provided herein may be modified by non-ligand group.
- non-ligand molecules have been conjugated to oligonucleotides in order to enhance the activity, cellular distribution, cellular targeting, or cellular uptake of the oligonucleotide, and procedures for performing such conjugations are available in the scientific literature.
- Such non-ligand moieties have included lipid moieties, such as cholesterol ( Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86:6553 ), arginine-rich peptides, cholic acid ( Manoharan et al., Bioorg. Med. Chem.
- a thioether e.g., hexyl-5-tritylthiol ( Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306 ; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3:2765 ), a thiocholesterol ( Oberhauser et al., Nucl.
- Acids Res., 1990, 18:3777 a polyamine or a polyethylene glycol chain ( Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969 ), or adamantane acetic acid ( Manoharan et al., Tetrahedron Lett., 1995, 36:3651 ), a palmityl moiety ( Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229 ), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety ( Crooke et al., J. Pharmacol. Exp.
- Typical conjugation protocols involve the synthesis of oligonucleotides bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction may be performed either with the oligonucleotide still bound to the solid support or following cleavage of the oligonucleotide in solution phase. Purification of the oligonucleotide conjugate by HPLC typically affords the pure conjugate.
- RNAi agents may be found in U.S. Application Publication Nos. 2007/0275465 , 2007/0054279 , 2006/0287260 , 2006/0035254 , 2006/0008822 . Also included are vector delivery systems that are capable of expressing the ArgRS-targeting sequences described herein. Included are vectors that express siRNA or other duplex-forming RNA interference molecules.
- a vector or nucleic acid construct system can comprise a single vector or plasmid, two or more vectors or plasmids, which together contain the total DNA to be introduced into the genome of the host cell, or a transposon.
- the choice of the vector will typically depend on the compatibility of the vector with the host cell into which the vector is to be introduced.
- the vector or nucleic acid construct is preferably one which is operably functional in a mammalian cell, such as a muscle cell.
- the vector can also include a selection marker such as an antibiotic or drug resistance gene, or a reporter gene (i.e., green fluorescent protein, luciferase), that can be used for selection or identification of suitable transformants or transfectants.
- Exemplary delivery systems may include viral vector systems (i.e ., viral-mediated transduction) including, but not limited to, retroviral (e.g., lentiviral) vectors, adenoviral vectors, adeno-associated viral vectors, and herpes viral vectors, among others known in the art.
- viral vector systems i.e ., viral-mediated transduction
- retroviral e.g., lentiviral
- adenoviral vectors e.g., adenoviral vectors
- adeno-associated viral vectors e.g., adeno-associated viral vectors
- herpes viral vectors e.g., herpes viral vectors
- Certain embodiments relate to the use of ArgRS polypeptides, antibodies, or polynucleotides in drug discovery, typically to identify agents that modulate one or more of the non-canonical activities of the reference ArgRS polypeptide, e.g., the ArgRS protein fragment.
- certain embodiments include methods of identifying one or more "cellular binding partners" of an ArgRS reference polypeptide, such as a cellular protein, lipid, nucleic acid or other host molecule that directly or physically interacts with the ArgRS polypeptide.
- Particular examples include for example cell-surface receptors, such as GPCRs, protein-protein interaction domains, and extracellular or intracellular domains thereof.
- host molecules include both upstream and downstream components of the non-canonical pathway, typically related by about 1, 2, 3, 4, 5 or more identifiable steps in the pathway, relative to the cellular binding partner/ArgRS protein interaction.
- Certain aspects include methods of identifying a compound (e.g., polypeptide) or other agent that agonizes or antagonizes the non-canonical activity of an ArgRS reference polypeptide or active variant thereof, such as by interacting with the ArgRS polypeptide and/or one or more of its cellular binding partners. Also included are methods of identifying agents that modulate the expression (e.g., splicing) of ArgRS splice variants, or modulate the activity of proteases that otherwise regulate the production of endogenous ArgRS protein fragments (resectins) at the protein level.
- a compound e.g., polypeptide
- agent that agonizes or antagonizes the non-canonical activity of an ArgRS reference polypeptide or active variant thereof, such as by interacting with the ArgRS polypeptide and/or one or more of its cellular binding partners.
- Certain embodiments therefore include methods of identifying a binding partner of an ArgRS reference polypeptide, comprising a) combining the ArgRS polypeptide with a biological sample under suitable conditions, and b) detecting specific binding of the ArgRS polypeptide to a binding partner, thereby identifying a binding partner that specifically binds to the ArgRS reference polypeptide.
- Also included are methods of screening for a compound that specifically binds to an ArgRS reference polypeptide or a binding partner of the ArgRS polypeptide comprising a) combining the polypeptide or the binding partner with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide or the binding partner to the test compound, thereby identifying a compound that specifically binds to the polypeptide or its binding partner.
- the compound is a polypeptide or peptide.
- the compound is a small molecule or other ( e.g., non-biological) chemical compound.
- the compound is a peptide mimetic.
- Any method suitable for detecting protein-protein interactions may be employed for identifying cellular proteins that interact with an ArgRS reference polypeptide, interact with one or more of its cellular binding partners, or both.
- traditional methods include co-immunoprecipitation, cross-linking, and co-purification through gradients or chromatographic columns of cell lysates or proteins obtained from cell lysates, mainly to identify proteins in the lysate that interact with the ArgRS polypeptide.
- amino acid sequence of a protein that interacts with an ArgRS polypeptide or its binding partner can be ascertained using techniques well known to those of skill in the art, such as via the Edman degradation technique. See, e.g., Creighton Proteins: Structures and Molecular Principles, W. H. Freeman & Co., N.Y., pp. 34 49, 1983 .
- the amino acid sequence obtained may be used as a guide for the generation of oligonucleotide mixtures that can be used to screen for gene sequences encoding such proteins. Screening may be accomplished, for example, by standard hybridization or PCR techniques, as described herein and known in the art.
- methods may be employed in the simultaneous identification of genes that encode the binding partner or other polypeptide. These methods include, for example, probing expression libraries, in a manner similar to the well known technique of antibody probing of lambda-gt11 libraries, using labeled ArgRS protein, or another polypeptide, peptide or fusion protein, e.g., a variant ArgRS polypeptide or ArgRS domain fused to a marker (e.g., an enzyme, fluor, luminescent protein, or dye), or an Ig-Fc domain.
- a marker e.g., an enzyme, fluor, luminescent protein, or dye
- plasmids may be constructed that encode two hybrid proteins: one plasmid consists of nucleotides encoding the DNA-binding domain of a transcription activator protein fused to an ArgRS reference nucleotide sequence (or, in certain embodiments, its binding partner), or a variant thereof, and the other plasmid consists of nucleotides encoding the transcription activator protein's activation domain fused to a cDNA (or collection of cDNAs) encoding an unknown protein(s) that has been recombined into the plasmid as part of a cDNA library.
- the DNA-binding domain fusion plasmid and the activator cDNA library may be transformed into a strain of the yeast Saccharomyces cerevisiae that contains a reporter gene (e.g., HBS or lacZ) whose regulatory region contains the transcription activator's binding site.
- a reporter gene e.g., HBS or lacZ
- the two-hybrid system or other such methodology may be used to screen activation domain libraries for proteins that interact with the "bait" gene product.
- an ArgRS reference polypeptide or variant may be used as the bait gene product.
- An ArgRS binding partner may also be used as a "bait” gene product.
- Total genomic or cDNA sequences are fused to the DNA encoding an activation domain.
- This library and a plasmid encoding a hybrid of a bait ArgRS gene product fused to the DNA-binding domain are co-transformed into a yeast reporter strain, and the resulting transformants are screened for those that express the reporter gene.
- a cDNA library of the cell line from which proteins that interact with bait ArgRS gene products are to be detected can be made using methods routinely practiced in the art.
- the cDNA fragments can be inserted into a vector such that they are translationally fused to the transcriptional activation domain of GAL4.
- This library can be co-transformed along with the bait gene-GAL4 fusion plasmid into a yeast strain, which contains a lacZ gene driven by a promoter that contains GAL4 activation sequence.
- a cDNA encoded protein, fused to GAL4 transcriptional activation domain, that interacts with bait gene product will reconstitute an active GAL4 protein and thereby drive expression of the HIS3 gene.
- Colonies which express HIS3, can be detected by their growth on Petri dishes containing semi-solid agar based media lacking histidine. The cDNA can then be purified from these strains, and used to produce and isolate the bait ArgRS gene-interacting protein using techniques routinely practiced in the art.
- RNA-protein interactions in yeast. See, e.g., Hook et al., RNA. 11:227-233, 2005 . Accordingly, these and related methods can be used to identify a cellular binding partner of an ArgRS polypeptide, and to identify other proteins or nucleic acids that interact with the ArgRS polypeptide, the cellular binding partner, or both.
- Certain embodiments relate to the use of interactome screening approaches.
- Particular examples include protein domain-based screening (see, e.g., Boxem et al., Cell. 134:534-545, 2008 ; and Yu et al., Science. 322:10-110, 2008 ).
- binding partners can be identified and can, in turn, be used in conjunction with standard techniques to identify proteins or other compounds with which it interacts.
- Certain embodiments thus relate to methods of screening for a compound that specifically binds to the binding partner of an ArgRS reference polypeptide, comprising a) combining the binding partner with at least one test compound under suitable conditions, and b) detecting binding of the binding partner to the test compound, thereby identifying a compound that specifically binds to the binding partner.
- the test compound is a polypeptide.
- the test compound is a chemical compound, such as a small molecule compound or peptide mimetic.
- Certain embodiments include methods of screening for a compound that modulates the activity of an ArgRS reference polypeptide, comprising a) combining the polypeptide with at least one test compound under conditions permissive for the activity of the polypeptide, b) assessing the activity of the polypeptide in the presence of the test compound, and c) comparing the activity of the polypeptide in the presence of the test compound with the activity of the polypeptide in the absence of the test compound, wherein a change in the activity of the polypeptide in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide.
- Certain embodiments include methods of screening for a compound that modulates the activity of a binding partner of an ArgRS reference polypeptide, comprising a) combining the polypeptide with at least one test compound under conditions permissive for the activity of the binding partner, b) assessing the activity of the binding partner in the presence of the test compound, and c) comparing the activity of the binding partner in the presence of the test compound with the activity of the binding partner in the absence of the test compound, wherein a change in the activity of the binding partner in the presence of the test compound is indicative of a compound that modulates the activity of the binding partner.
- these and related embodiments include assessing a selected non-canonical activity that is associated with the ArgRS polypeptide or its binding partner. Included are in vitro and in vivo conditions, such as cell culture conditions.
- Certain embodiments include methods of screening a compound for effectiveness as a full or partial agonist of an ArgRS reference polypeptide or an active fragment or variant thereof, comprising a) exposing a sample comprising the polypeptide to a compound, and b) detecting agonist activity in the sample, typically by measuring an increase in the non-canonical activity of the ArgRS polypeptide. Certain methods include a) exposing a sample comprising a binding partner of the ArgRS polypeptide to a compound, and b) detecting agonist activity in the sample, typically by measuring an increase in the selected non-canonical activity of the ArgRS polypeptide. Certain embodiments include compositions that comprise an agonist compound identified by the method and a pharmaceutically acceptable carrier or excipient.
- compositions that comprise an antagonist compound identified by the method and a pharmaceutically acceptable carrier or excipient.
- in vitro systems may be designed to identify compounds capable of interacting with or modulating an ArgRS reference sequence or its binding partner.
- Certain of the compounds identified by such systems may be useful, for example, in modulating the activity of the pathway, and in elaborating components of the pathway itself. They may also be used in screens for identifying compounds that disrupt interactions between components of the pathway; or may disrupt such interactions directly.
- One exemplary approach involves preparing a reaction mixture of the ArgRS polypeptide and a test compound under conditions and for a time sufficient to allow the two to interact and bind, thus forming a complex that can be removed from and/or detected in the reaction mixture
- an ArgRS polypeptide, a cellular binding partner, or test compound(s) can be anchored onto a solid phase.
- the resulting complexes may be captured and detected on the solid phase at the end of the reaction.
- the ArgRS polypeptide and/or its binding partner are anchored onto a solid surface, and the test compound(s), which are not anchored, may be labeled, either directly or indirectly, so that their capture by the component on the solid surface can be detected.
- test compound(s) are anchored to the solid surface, and the ArgRS polypeptide and/or its binding partner, which are not anchored, are labeled or in some way detectable.
- microtiter plates may conveniently be utilized as the solid phase.
- the anchored component (or test compound) may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished by simply coating the solid surface with a solution of the protein and drying.
- an immobilized antibody preferably a monoclonal antibody, specific for the protein to be immobilized may be used to anchor the protein to the solid surface.
- the surfaces may be prepared in advance and stored.
- the non-immobilized component is typically added to the coated surface containing the anchored component. After the reaction is complete, un-reacted components are removed ( e.g., by washing) under conditions such that any specific complexes formed will remain immobilized on the solid surface.
- the detection of complexes anchored on the solid surface can be accomplished in a number of ways. For instance, where the previously non-immobilized component is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed.
- an indirect label can be used to detect complexes anchored on the surface; e.g ., using a labeled antibody specific for the previously non-immobilized component (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).
- the presence or absence of binding of a test compound can be determined, for example, using surface plasmon resonance (SPR) and the change in the resonance angle as an index, wherein an ArgRS polypeptide or a cellular binding partner is immobilized onto the surface of a commercially available sensorchip (e.g., manufactured by BIACORE TM ) according to a conventional method, the test compound is contacted therewith, and the sensorchip is illuminated with a light of a particular wavelength from a particular angle.
- SPR surface plasmon resonance
- the binding of a test compound can also be measured by detecting the appearance of a peak corresponding to the test compound by a method wherein an ArgRS polypeptide or a cellular binding partner is immobilized onto the surface of a protein chip adaptable to a mass spectrometer, a test compound is contacted therewith, and an ionization method such as MALDI-MS, ESI-MS, FAB-MS and the like is combined with a mass spectrometer (e.g., double-focusing mass spectrometer, quadrupole mass spectrometer, time-of-flight mass spectrometer, Fourier transformation mass spectrometer, ion cyclotron mass spectrometer and the like).
- a mass spectrometer e.g., double-focusing mass spectrometer, quadrupole mass spectrometer, time-of-flight mass spectrometer, Fourier transformation mass spectrometer, ion cyclotron mass spectrometer and the like.
- cell-based assays, membrane vesicle-based assays, or membrane fraction-based assays can be used to identify compounds that modulate interactions in the non-canonical pathway of the selected ArgRS polypeptide.
- cell lines that express an ArgRS polypeptide and/or a binding partner, or a fusion protein containing a domain or fragment of such proteins (or a combination thereof), or cell lines ( e.g., COS cells, CHO cells, HEK293 cells, Hela cells etc.) that have been genetically engineered to express such protein(s) or fusion protein(s) can be used.
- Test compound(s) that influence the non-canonical activity can be identified by monitoring a change (e.g., a statistically significant change) in that activity as compared to a control or a predetermined amount.
- RNAi agents for example, also included are methods of screening a compound for effectiveness in altering expression of an ArgRS reference polynucleotide, comprising a) exposing a sample comprising the ArgRS reference polynucleotide to a compound such as a potential antisense oligonucleotide, and b) detecting altered expression of the ArgRS polynucleotide.
- these and related embodiments can be employed in cell-based assays or in cell-free translation assays, according to routine techniques in the art.
- the antisense and RNAi agents identified by such methods.
- Antibodies to ArgRS protein fragments can also be used in screening assays, such as to identify an agent that specifically binds to an ArgRS, confirm the specificity or affinity of an agent that binds to an ArgRS protein fragment, or identify the site of interaction between the agent and the ArgRS protein fragment. Included are assays in which the antibody is used as a competitive inhibitor of the agent. For instance, an antibody that specifically binds to an ArgRS protein fragment with a known affinity can act as a competitive inhibitor of a selected agent, and be used to calculate the affinity of the agent for the ArgRS protein fragment. Also, one or more antibodies that specifically bind to known epitopes or sites of an ArgRS protein fragment can be used as a competitive inhibitor to confirm whether or not the agent binds at that same site. Other variations will be apparent to persons skilled in the art.
- HTS high-throughput screening
- HTS typically uses automation to run a screen of an assay against a library of candidate compounds, for instance, an assay that measures an increase or a decrease in a non-canonical activity, as described herein.
- Any of the screening methods provided herein may utilize small molecule libraries or libraries generated by combinatorial chemistry.
- Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention. Examples of methods for the synthesis of molecular libraries can be found in: (Carell et al., 1994a; Carell et al., 1994b; Cho et al., 1993; DeWitt et al., 1993; Gallop et al., 1994; Zuckermann et al., 1994).
- Libraries of compounds may be presented in solution (Houghten et al., 1992) or on beads (Lam et al., 1991), on chips (Fodor et al., 1993), bacteria, spores ( Ladner et al., U.S. Pat. No. 5,223,409, 1993 ), plasmids (Cull et al., 1992) or on phage (Cwirla et al., 1990; Devlin et al., 1990; Felici et al., 1991; Ladner et al., U.S. Pat. No. 5,223,409, 1993 ; Scott and Smith, 1990).
- Embodiments of the present invention encompass the use of different libraries for the identification of small molecule modulators of one or more ArgRS protein fragments, their cellular binding partners, and/or their related non-canonical activities.
- Libraries useful for the purposes of the invention include, but are not limited to, (1) chemical libraries, (2) natural product libraries, and (3) combinatorial libraries comprised of random peptides, oligonucleotides and/or organic molecules.
- Chemical libraries consist of structural analogs of known compounds or compounds that are identified as “hits” or “leads” via natural product screening.
- Natural product libraries are derived from collections of microorganisms, animals, plants, or marine organisms which are used to create mixtures for screening by: (1) fermentation and extraction of broths from soil, plant or marine microorganisms or (2) extraction of plants or marine organisms.
- Natural product libraries include polyketides, non-ribosomal peptides, and variants (non-naturally occurring) thereof. See, e.g., Cane et al., Science 282:63-68, 1998 .
- Combinatorial libraries may be composed of large numbers of peptides, oligonucleotides or organic compounds as a mixture. They are relatively easy to prepare by traditional automated synthesis methods, PCR, cloning or proprietary synthetic methods.
- a combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis, by combining a number of chemical "building blocks” such as reagents.
- a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks (amino acids) in every possible way for a given compound length ( i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks.
- Embodiments of the present invention include therapeutic methods of treatment.
- the ArgRS agents described herein including ArgRS polypeptides, ArgRS polynucleotides, ArgRS polynucleotide-based vectors, ArgRS expressing host cells, antisense oligonucleotides, RNAi agents, as well as binding agents such as peptides, antibodies and antigen-binding fragments, peptide mimetics and other small molecules, can be used to treat a variety of non-limiting diseases or conditions associated with the non-canonical activities of a reference ArgRS.
- non-canonical activities include modulation of extracellular signaling, modulation of cell proliferation, modulation of cell migration, modulation of cell differentiation (e.g., hematopoiesis, neurogenesis, myogenesis, osteogenesis, and adipogenesis), modulation of apoptosis or other forms of cell death, modulation of angiogenesis, modulation of cell binding, modulation of cellular metabolism, modulation of cytokine production or activity, modulation of cytokine receptor activity, modulation of cellular uptake, or secretion, immunomodulation, modulation of inflammation, modulation of metabolic processes such as glucose control, and the like.
- modulation of extracellular signaling modulation of cell proliferation, modulation of cell migration, modulation of cell differentiation (e.g., hematopoiesis, neurogenesis, myogenesis, osteogenesis, and adipogenesis), modulation of apoptosis or other forms of cell death
- modulation of angiogenesis modulation of cell binding
- modulation of cellular metabolism modulation of cyto
- polynucleotide-based therapies such as antisense therapies and RNAi interference therapies, which typically relate to reducing the expression of a target molecule, such as an endogenous fragment of an ArgRS, or a cellular binding partner of an ArgRS polypeptide, which otherwise contributes to its non-canonical activity.
- Antisense or RNAi therapies typically antagonize the non-canonical activity, such as by reducing expression of the ArgRS reference polypeptide.
- polypeptides or peptides include antibodies or antigen-binding fragment, peptide mimetics, or other small molecule-based therapies, which either agonize or antagonize the non-canonical activity of an ArgRS reference polypeptide, such as by interacting directly with the ArgRS polypeptide, its cellular binding partner(s), or both.
- ArgRS agents or compositions of the present invention include methods of using the ArgRS agents or compositions of the present invention for treating a cell, tissue or subject.
- the cells or tissues that may be treated or modulated by the present invention are preferably mammalian cells or tissues, or more preferably human cells or tissues. Such cells or tissues can be of a healthy state or of a diseased state.
- methods for modulating therapeutically relevant cellular activities including, but not limited to, cellular metabolism, cell differentiation, cell proliferation, cellular uptake, cell secretion, cell death, cell mobilization, cell migration, gene transcription, mRNA translation, cell impedance, immune responses, inflammatory responses, and the like, comprising contacting a cell with an ArgRS agent or composition as described herein.
- the cell is in a subject.
- the ArgRS compositions may be employed in treating essentially any cell or tissue or subject that would benefit from modulation of one or more such activities.
- ArgRS agents and compositions may also be used in any of a number of therapeutic contexts including, for example, those relating to the treatment or prevention of neoplastic diseases, immune system diseases or conditions (e.g., autoimmune diseases and inflammation), infectious diseases, metabolic diseases, neuronal/neurological diseases, muscular/cardiovascular diseases, diseases associated with aberrant hematopoiesis, diseases associated with aberrant myogenesis, diseases associated with aberrant neurogenesis, diseases associated with aberrant adipogenesis, diseases associated with aberrant osteogenesis, diseases associated with aberrant angiogenesis, diseases associated with aberrant cell survival, diseases associated with aberrant lipid uptake, diseases associated with aging (e.g. hearing loss, peripheral or autonomic neuropathies, senile dementia, retinopathy) and others.
- neoplastic diseases e.g., immune system diseases or conditions
- infectious diseases e.g., metabolic diseases, neuronal/neurological diseases, muscular/cardiovascular diseases, diseases associated with aberrant hematopoiesis,
- the ArgRS compositions of the invention may be used to modulate angiogenesis, e.g ., via modulation of endothelial cell proliferation and/or signaling.
- Endothelial cell proliferation and/or signaling may be monitored using an appropriate cell line (e.g., human microvascular endothelial lung cells (HMVEC-L) and human umbilical vein endothelial cells (HUVEC)), and using an appropriate assay (e.g., endothelial cell migration assays, endothelial cell proliferation assays, tube-forming assays, matrigel plug assays, etc.), many of which are known and available in the art.
- HMVEC-L human microvascular endothelial lung cells
- HUVEC human umbilical vein endothelial cells
- an appropriate assay e.g., endothelial cell migration assays, endothelial cell proliferation assays, tube-forming assays, matrigel plug assays, etc.
- the compositions of the invention may be employed in the treatment of essentially any cell or tissue or subject that would benefit from modulation of angiogenesis.
- a cell or tissue or subject experiencing or susceptible to angiogenesis e.g., an angiogenic condition
- a suitable composition of the invention to inhibit an angiogenic condition.
- a cell or tissue experiencing or susceptible to insufficient angiogenesis e.g., an angiostatic condition
- hematopoietic processes that may be modulated by the ArgRS polypeptides of the invention include, without limitation, the formation of myeloid cells (e.g., erythroid cells, mast cells monocytes/macrophages, myeloid dendritic cells, granulocytes such as basophils, neutrophils, and eosinophils, megakaryocytes, platelets) and lymphoid cells (e.g., natural killer cells, lymphoid dendritic cells, B-cells, and T-cells).
- myeloid cells e.g., erythroid cells, mast cells monocytes/macrophages, myeloid dendritic cells, granulocytes such as basophils, neutrophils, and eosinophils, megakaryocytes, platelets
- lymphoid cells e.g., natural killer cells, lymphoid dendritic cells, B-cells, and T-cells.
- Certain specific hematopoietic processes include erythropoiesis, granulopoiesis, lymphopoiesis, megakaryopoiesis, thrombopoiesis, and others. Also included are methods of modulating the trafficking or mobilization of hematopoietic cells, including hematopoietic stem cells, progenitor cells, erythrocytes, granulocytes, lymphocytes, megakaryocytes, and thrombocytes.
- the methods of modulating hematopoiesis may be practiced in vivo, in vitro, ex vivo, or in any combination thereof. These methods can be practiced on any biological sample, cell culture, or tissue that contains hematopoietic stem cells, hematopoietic progenitor cells, or other stem or progenitor cells that are capable of differentiating along the hematopoietic lineage (e.g., adipose tissue derived stem cells).
- stem cells and progenitor cells whether of hematopoietic origin or otherwise, can be isolated and/or identified according to the techniques and characteristics described herein and known in the art.
- compositions of the invention may also be useful as immunomodulators for treating anti- or pro-inflammatory indications by modulating the cells that mediate, either directly or indirectly, autoimmune and/or inflammatory diseases, conditions and disorders.
- the utility of the compositions of the invention as immunomodulators or modulators of inflammation can be monitored using any of a number of known and available techniques in the art including, for example, migration assays (e.g., using leukocytes or lymphocytes) or cell viability assays (e.g., using B-cells, T-cells, monocytes or NK cells).
- Inflammation refers generally to the biological response of tissues to harmful stimuli, such as pathogens, damaged cells ( e.g., wounds), and irritants.
- the term "inflammatory response” refers to the specific mechanisms by which inflammation is achieved and regulated, including, merely by way of illustration, immune cell activation or migration, cytokine production, vasodilation, including kinin release, fibrinolysis, and coagulation, among others described herein and known in the art.
- Chronic inflammation is associated with a variety of pathological conditions or diseases, including, for example, allergies, Alzheimer's disease, anemia, aortic valve stenosis, arthritis such as rheumatoid arthritis and osteoarthritis, cancer, congestive heart failure, fibromyalgia, fibrosis, heart attack, kidney failure, lupus, pancreatitis, stroke, surgical complications, inflammatory lung disease, inflammatory bowel disease, atherosclerosis, neurological disorders, diabetes, metabolic disorders, obesity, and psoriasis, among others described herein and known in the art.
- ArgRS compositions may be used to treat or manage chronic inflammation, modulate any of one or more of the individual chronic inflammatory responses, or treat any one or more diseases or conditions associated with chronic inflammation.
- Criteria for assessing the signs and symptoms of inflammatory and other conditions including for purposes of making differential diagnosis and also for monitoring treatments such as determining whether a therapeutically effective dose has been administered in the course of treatment, e.g ., by determining improvement according to accepted clinical criteria, will be apparent to those skilled in the art and are exemplified by the teachings of e.g., Berkow et al., eds., The Merck Manual, 16th edition, Merck and Co., Rahway, N.J., 1992 ; Goodman et al., eds., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th edition, Pergamon Press, Inc., Elmsford, N.Y., (2001 ); Avery's Drug Treatment: Principles and Practice of Clinical Pharmacology and Therapeutics, 3rd edition, ADIS Press, Ltd., Williams and Wilkins, Baltimore, MD.
- the ArgRS compositions of the invention may be used to modulate cellular proliferation and/or survival and, accordingly, for treating or preventing diseases, disorders or conditions characterized by abnormalities in cellular proliferation and/or survival.
- the ArgRS compositions may be used to modulate apoptosis and/or to treat diseases or conditions associated with abnormal apoptosis.
- Apoptosis can be monitored by any of a number of available techniques known and available in the art including, for example, assays that measure fragmentation of DNA, alterations in membrane asymmetry, activation of apoptotic caspases and/or release of cytochrome C and AIF.
- Embodiments of the present invention include ArgRS polynucleotides, ArgRS polypeptides, host cells expressing ArgRS polypeptides, binding agents, modulatory agents, or other compounds described herein, formulated in pharmaceutically-acceptable or physiologically-acceptable solutions for administration to a cell or an animal, either alone, or in combination with one or more other modalities of therapy. It will also be understood that, if desired, the compositions of the invention may be administered in combination with other agents as well, such as, e.g., other proteins or polypeptides or various pharmaceutically-active agents. There is virtually no limit to other components that may also be included in the compositions, provided that the additional agents do not adversely affect the modulatory or other effects desired to be achieved.
- compositions of the invention formulation of pharmaceutically-acceptable excipients and carrier solutions is well-known to those of skill in the art, as is the development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g ., oral, parenteral, intravenous, intranasal, subcutaneous, and intramuscular administration and formulation.
- the pharmaceutical or therapeutic compositions of the invention do not stimulate an immune reaction.
- the pharmaceutical or therapeutic compositions of the invention typically comprising one or more ArgRS polypeptides or polynucleotides, stimulate an immune reaction, such as by serving as an adjuvant in a vaccine or related composition, or being present in a composition together with a separate adjuvant or agent stimulates an immune response.
- the ArgRS agents such as ArgRS polypeptides, ArgRS polynucleotides, and antibodies have a solubility that is desirable for the particular mode of administration, such intravenous administration.
- desirable solubilities include at least about 1 mg/ml, at least about 10 mg/ml, at least about 25 mg/ml, and at least about 50 mg/ml.
- compositions disclosed herein may be delivered via oral administration to a subject.
- these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
- compositions disclosed herein parenterally, subcutaneously, intravenously, intramuscularly, intra-arterially, intrathecally, intraparenchymally, intraventricularly, intraurethrally, intrasternally, intracranially, intrasynovially, or even intraperitoneally as described, for example, in U.S. Pat. No. 5,543,158 ; U.S. Pat. No. 5,641,515 and U.S. Pat. No. 5,399,363 .
- Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors, and infusion techniques.
- Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
- the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions ( U.S. Pat. No. 5,466,468 ).
- the form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
- the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol ( e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
- Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
- a coating such as lecithin
- surfactants for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
- isotonic agents for example, sugars or sodium chloride.
- Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
- aqueous solution for parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
- aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
- a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure.
- one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion (see, e.g., Remington's Pharmaceutical Sciences, 15th Edition, pp.
- Sterile injectable solutions can be prepared by incorporating the active compounds in the required amount in the appropriate solvent with the various other ingredients enumerated above, as required, followed by filtered sterilization.
- dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
- compositions disclosed herein may be formulated in a neutral or salt form.
- Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
- solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
- the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug-release capsules, and the like.
- carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
- carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
- the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
- compositions that do not produce an allergic or similar untoward reaction when administered to a human.
- pharmaceutically-acceptable refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.
- aqueous composition that contains a protein as an active ingredient is well understood in the art.
- injectables either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared.
- the preparation can also be emulsified.
- the pharmaceutical compositions may be delivered by intranasal sprays, inhalation, and/or other aerosol delivery vehicles.
- Methods for delivering genes, polynucleotides, and peptide compositions directly to the lungs via nasal aerosol sprays have been described e.g ., in U.S. Pat. No. 5,756,353 and U.S. Pat. No. 5,804,212 .
- the delivery of drugs using intranasal microparticle resins Takenaga et al., 1998) and lysophosphatidyl-glycerol compounds ( U.S. Pat. No. 5,725,871 ) are also well-known in the pharmaceutical arts.
- transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 .
- compositions may be formulated to be immediate and / or sustained release.
- Sustained release compositions include delayed, modified, pulsed, controlled, targeted and programmed release.
- the compositions may be formulated as a suspension or as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing sustained release of the ArgRS polynucleotides, ArgRS polypeptides, binding agents, modulatory agents and other active agents.
- formulations include without limitation, drug-coated stents and semi-solids and suspensions comprising drug-loaded poly(DL-lactic-co-glycolic)acid (PGLA), poly(DL-lactide-co-glycolide) (PLG) or poly(lactide) (PLA) lamellar vesicles or microparticles, hydrogels ( Hoffman AS: Ann. N.Y. Acad. Sci.
- PGLA poly(DL-lactic-co-glycolic)acid
- PLA poly(DL-lactide-co-glycolide)
- PLA poly(lactide) lamellar vesicles or microparticles
- hydrogels Hoffman AS: Ann. N.Y. Acad. Sci.
- poly-amino acid nanoparticles systems sold under the trademark MEDUSA® developed by Flamel Technologies Inc.
- non aqueous gel systems sold under the trademark ATRIGEL® developed by Atrix, Inc.
- Sucrose Acetate Isobutyrate Extended Release formulations sold under the trademark SABER® developed by Durect Corporation
- lipid-based systems developed by SkyePharma and sold under the trademark DEPOFOAM®.
- Sustained release devices capable of delivering desired doses of the pharmaceutical compositions over extended periods of time are known in the art.
- US Pat. Nos. 5,034,229 ; 5,557,318 ; 5,110,596 ; 5,728,396 ; 5,985,305 ; 6,113,938 ; 6,156,331 ; 6,375,978 ; and 6,395,292 teach osmotically-driven devices capable of delivering an active agent formulation, such as a solution or a suspension, at a desired rate over an extended period of time (i.e., a period ranging from more than one week up to one year or more).
- exemplary sustained release devices include regulator-type pumps that provide constant flow, adjustable flow, or programmable flow of beneficial agent formulations, which are available from Medtronic including the Intrathecal pumps sold under the trademark SYNCHROMED INFUSION SYSTEM ® , the Johnson and Johnson systems sold under the trademark CODMAN ® division pumps, and INSET ® technologies pumps. Further examples of devices are described in US Pat. Nos. 6,283,949 ; 5,976,109 ; 5,836,935 ; and 5,511,355 .
- the delivery may occur by use of liposomes, nanocapsules, microparticles, microspheres, lipid particles, vesicles, and the like, for the introduction of the compositions of the present invention into suitable host cells.
- the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, a nanoparticle or the like.
- the formulation and use of such delivery vehicles can be carried out using known and conventional techniques.
- the agents provided herein may be attached to a pharmaceutically acceptable solid substrate, including biocompatible and biodegradable substrates such as polymers and matrices.
- solid substrates include, without limitation, polyesters, hydrogels (for example, poly(2-hydroxyethylmethacrylate), or poly(vinylalcohol)), polylactides ( U.S. Pat. No.
- copolymers of L-glutamic acid and ⁇ -ethyl-L-glutamate copolymers of L-glutamic acid and ⁇ -ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as poly(lactic-co-glycolic acid) (PLGA) and the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), poly-D-(-)-3-hydroxybutyric acid, collagen, metal, hydroxyapatite, bioglass, aluminate, bioceramic materials, and purified proteins.
- PLGA poly(lactic-co-glycolic acid)
- LUPRON DEPOTTM injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate
- the solid substrate comprises biodegradable polymers sold under the trademark ATRIGELTM (QLT, Inc., Vancouver, B.C.).
- ATRIGEL ® drug delivery system consists of biodegradable polymers dissolved in biocompatible carriers. Pharmaceuticals may be blended into this liquid delivery system at the time of manufacturing or, depending upon the product, may be added later by the physician at the time of use.
- the liquid product is injected into the subcutaneous space through a small gauge needle or placed into accessible tissue sites through a cannula, water in the tissue fluids causes the polymer to precipitate and trap the drug in a solid implant. The drug encapsulated within the implant is then released in a controlled manner as the polymer matrix biodegrades with time.
- compositions for use in the present invention may also be administered topically, (intra)dermally, or transdermally to the skin or mucosa.
- Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibers, bandages, and microemulsions.
- Liposomes may also be used.
- Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol, and propylene glycol.
- Penetration enhancers may be incorporated-see, e.g., Finnin and Morgan: J. Pharm. Sci.
- Topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis, and microneedle or needle-free injection for example using the systems sold under the trademarks POWDERJECTTM, and BIOJECTTM.
- compositions and agents provided herein may be administered according to the methods of the present invention in any therapeutically effective dosing regimen.
- the dosage amount and frequency are selected to create an effective level of the agent without harmful effects.
- the effective amount of a compound of the present invention will depend on the route of administration, the type of warm-blooded animal being treated, and the physical characteristics of the specific warm-blooded animal under consideration. These factors and their relationship to determining this amount are well known to skilled practitioners in the medical arts. This amount and the method of administration can be tailored to achieve optimal efficacy but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize.
- the amount of a composition or agent administered will generally range from a dosage of from about 0.1 to about 100 mg/kg/day, and typically from about 0.1 to 10 mg/kg where administered orally or intravenously.
- a dosage is 5 mg/kg or 7.5 mg/kg.
- the dosage is about 50-2500 mg per day, 100-2500 mg/day, 300-1800 mg/day, or 500-1800 mg/day.
- the dosage is between about 100 to 600 mg/day.
- the dosage is between about 300 and 1200 mg/day.
- the composition or agent is administered at a dosage of 100 mg/day, 240 mg/day 300 mg/day, 600 mg/day, 1000 mg/day, 1200 mg/day, or 1800 mg/day, in one or more doses per day ( i.e., where the combined doses achieve the desired daily dosage).
- a dosage is 100 mg bid, 150 mg bid, 240 mg bid, 300 mg bid, 500 mg bid, or 600 mg bid.
- the composition or agent is administered in single or repeat dosing. The initial dosage and subsequent dosages may be the same or different.
- a composition or agent is administered in a single dosage of 0.1 to 10 mg/kg or 0.5 to 5 mg/kg. In other embodiments, a composition or agent is administered in a dosage of 0.1 to 50 mg/kg/day, 0.5 to 20 mg/kg/day, or 5 to 20 mg/kg/day.
- a composition or agent is administered orally or intravenously, e.g., by infusion over a period of time of about, e.g., 10 minutes to 90 minutes.
- a composition or agent is administered by continuous infusion, e.g ., at a dosage of between about 0.1 to about 10 mg/kg/hr over a time period. While the time period can vary, in certain embodiments the time period may be between about 10 minutes to about 24 hours or between about 10 minutes to about three days.
- an effective amount or therapeutically effective amount is an amount sufficient to achieve a total concentration of the composition or agent in the blood plasma of a subject with a C max of between about 0.1 ⁇ g/ml and about 20 ⁇ g/ml or between about 0.3 ⁇ g/ml and about 20 ⁇ g/ml.
- an oral dosage is an amount sufficient to achieve a blood plasma concentration (C max ) of between about 0.1 ⁇ g/ml to about 5 ⁇ g/ml or between about 0.3 ⁇ g/ml to about 3 ⁇ g/ml.
- an intravenous dosage is an amount sufficient to achieve a blood plasma concentration (C max ) of between about 1 ⁇ g/ml to about 10 ⁇ g/ml or between about 2 ⁇ g/ml and about 6 ⁇ g/ml.
- C max blood plasma concentration
- the total concentration of an agent in the blood plasma of the subject has a mean trough concentration of less than about 20 ⁇ g/ml and/or a steady state concentration of less than about 20 ⁇ g/ml.
- the total concentration of an agent in the blood plasma of the subject has a mean trough concentration of less than about 10 ⁇ g/ml and/or a steady state concentration of less than about 10 ⁇ g/ml.
- the total concentration of an agent in the blood plasma of the subject has a mean trough concentration of between about 1 ng/ml and about 10 ⁇ g/ml and/or a steady state concentration of between about 1 ng/ml and about 10 ⁇ g/ml. In one embodiment, the total concentration of an agent in the blood plasma of the subject has a mean trough concentration of between about 0.3 ⁇ g/ml and about 3 ⁇ g/ml and/or a steady state concentration of between about 0.3 ⁇ g/ml and about 3 ⁇ g/ml.
- a composition or agent is administered in an amount sufficient to achieve in the mammal a blood plasma concentration having a mean trough concentration of between about 1 ng/ml and about 10 ⁇ g/ml and/or a steady state concentration of between about 1 ng/ml and about 10 ⁇ g/ml.
- the total concentration of the agent in the blood plasma of the mammal has a mean trough concentration of between about 0.3 ⁇ g/ml and about 3 ⁇ g/ml and/or a steady state concentration of between about 0.3 ⁇ g/ml and about 3 ⁇ g/ml.
- the effective amount of a composition or agent, or the blood plasma concentration of composition or agent is achieved or maintained, e.g ., for at least 15 minutes, at least 30 minutes, at least 45 minutes, at least 60 minutes, at least 90 minutes, at least 2 hours, at least 3 hours, at least 4 hours, at least 8 hours, at least 12 hours, at least 24 hours, at least 48 hours, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least one week, at least 2 weeks, at least one month, at least 2 months, at least 4 months, at least 6 months, at least one year, at least 2 years, or greater than 2 years.
- the amount of polypeptide administered will typically be in the range of about 0.1 ⁇ g/kg to about 0.1 mg/kg to about 50 mg/kg of patient body weight.
- about 0.1 ⁇ g/kg to about 0.1 mg/kg to about 50 mg/kg body weight (e.g., about 0.1-15 mg/kg/dose) of polypeptide can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
- a dosing regimen may comprise administering an initial loading dose of about 4 mg/kg, followed by a weekly maintenance dose of about 2 mg/kg of the polypeptide, or about half of the loading dose.
- a typical daily dosage might range from about 0.1 ⁇ g/kg to about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
- the treatment is sustained until a desired suppression of disease symptoms occurs.
- the effective dosage achieves the blood plasma levels or mean trough concentration of a composition or agent described herein. These may be readily determined using routine procedures.
- kits comprising one or more containers filled with one or more of the polypeptides, polynucleotides, antibodies, multiunit complexes, compositions thereof, etc., of the invention, as described herein.
- the kits can include written instructions on how to use such compositions (e.g., to modulate cellular signaling, angiogenesis, cancer, inflammatory conditions, diagnosis etc .) .
- kits herein may also include a one or more additional therapeutic agents or other components suitable or desired for the indication being treated, or for the desired diagnostic application.
- An additional therapeutic agent may be contained in a second container, if desired.
- additional therapeutic agents include, but are not limited to anti-neoplastic agents, anti-inflammatory agents, antibacterial agents, antiviral agents, angiogenic agents, etc.
- kits herein can also include one or more syringes or other components necessary or desired to facilitate an intended mode of delivery (e.g., stents, implantable depots, etc .).
- Polynucleotide sequences encoding epitope tagged versions of the ArgRS polypeptides were codon optimized and cloned into bacterial expression vectors using the methods listed below.
- E. coli codon-optimized DNA ( Welch et al., PLoS ONE 4(9): e7007 doi:10.1371/journal.pone.0007002 ) encoding each ArgRS polypeptide is synthesized by DNA 2.0 (Menlo Park, CA), and two versions of each ArgRS polypeptide are synthesized, containing either an N-terminal, or C-terminal combined epitope tag comprising both a six histidine tag and V5 epitope tag.
- DNA encoding the N-terminally tagged ArgRS polypeptides is synthesized with a 5' extension encoding in 5' to 3' orientation, a ribosome binding site (rbs (underlined below)), NdeI restriction site, six histidine tag, and a V5 epitope tag, (AGGAGGTAAAACAT ATGCATCATCATCATCATCACGGTAAGCCTATCCCTA ACCCTTTGCTCGGTCTCGATTCTACG) ( SEQ . ID. No. 1 ), which is fused in frame to the predicted ArgRS polypeptide open reading frame.
- ArgRS polypeptide comprises a predicted native initiation methionine (ATG) residue, or the first amino acid residue of the predicted ArgRS polypeptide is Met, this was deleted.
- ATG native initiation methionine
- Met the first amino acid residue of the predicted ArgRS polypeptide is Met
- TAGACTCGAG the first amino acid residue of the predicted ArgRS polypeptide is Met
- DNA encoding the C-terminally tagged ArgRS polypeptides is synthesized with a 5' extension encoding a rbs (underlined below) and NdeI restriction site that either recapitulates the predicted native start codon for the ArgRS polypeptide, or inserts an ATG in frame with the predicted ArgRS polypeptide open reading frame, (AGGAGATAAAACATATG) (SEQ. ID. No. 3).
- the ribosome binding site can comprise the sequences "AGGAGGTAAAACAT" ( SEQ . ID. No. 4), "AGGAGATAAAACAT” ( SEQ . ID. No. 5), or GAAGGAGATATACAT ( SEQ . ID. No. 6).
- a 3' extension is synthesized which encodes in 5' to 3' order, a V5 epitope tag, six histidine tag, two stop codons and a XhoI site, (GGTAAGCCTATCCCTAACCCTCTCCTCGGTCTCGATTCTACGCACCACCATC ATCACCATTAATGACTCGAG) (SEQ. ID. No. 7), which is fused in frame to the predicted ArgRS polypeptide open reading frame. If the ArgRS polypeptide included a predicted native stop codon, this was deleted.
- E. coli codon-optimized DNA ( Ermolaeva MD (2001) Curr. Iss. Mol. Biol. 3 (4) 91-7 ) encoding each ArgRS polypeptide is synthesized by GENEWIZ (South Plainfield, NJ). Each polynucleotide sequence encoding the ArgRS polypeptide was synthesized with short 5' and 3' extensions comprising unique restriction sites for subsequent cloning.
- ArgRS polypeptide comprises a predicted native initiation methionine residue (ATG), or the first amino acid residue of the predicted ArgRS polypeptide is Met, this was deleted.
- a XhoI restriction site was inserted at the 3' end of the predicted open reading frame. In cases where the ArgRS polypeptide comprises a predicted native stop codon, this was deleted.
- the DNA encoding the N-tagged ArgRS polypeptide is cloned into the N-tagged vector (pET24b_N-6XHis/V5), which comprises a 5' DNA sequence encoding six histidines and a V5 epitope tag, (CATATGCATCATCATCATCATCATCACGGTAAGCCTATCCCTAACCCTCTCCTCGGTCTCGATTC TACGGGATCC) (SEQ. ID. No. 8), in frame with an initiation codon (ATG) embedded within the NdeI restriction site.
- This 5' extension is fused to the predicted ArgRS polypeptide open reading frame through a short 2 amino acid linker (GS).
- the DNA encoding the N-tagged ArgRS polypeptide comprises a DNA sequence encoding a 2 amino acid extension (LE) followed by two termination codons (CTCGAGTAATGA) ( SEQ . ID. No. 9).
- the DNA encoding the C-tagged ArgRS polypeptide cloned into the C-tagged vector comprises a 5' sequence encoding an initiation codon (ATG) embedded within the NdeI restriction site which is fused to the predicted ArgRS polypeptide open reading frame through a short 2 amino acid linker (GS), (CATATGGGATCC) (SEQ. ID. No. 10).
- the DNA encoding the C-tagged ArgRS polypeptide comprises a 3' DNA sequence encoding a short linker 2 amino acid linker (LE) followed by a V5 epitope tag followed by six histidines, and two stop codons,
- ArgRS polypeptides are expressed in bacteria in a medium-throughput format and/or in larger scale flask cultures depending upon the amount of protein required. ArgRS polypeptides are purified using affinity and ion exchange chromatography as described below, and as specified for specific experiments.
- Bacterial cuftures 100 ng of expression vector comprising codon optimized DNA encoding each ArgRS polypeptide (as described above) is transformed into BL21(DE3) (EMD chemicals, cat. no. 69450) competent E. coli bacteria at 42°C for 30 seconds in PCR plates. C41(DE3) (Lucigen, cat. no. 60442), HMS174(DE3) (EMD chemicals, cat. no. 69453) and Origami2(DE3) (EMD chemicals, cat. no. 71345) strains are also evaluated. The plates are placed on ice for 2 minutes and 100 ⁇ L of SOC medium is added, followed by a 1-hour incubation at 37°C. 5 mL of auto-induction medium (EMD chemicals, cat.
- Protein Isolation After the culture reached stationary phase (typical OD 600 of 3-6), the blocks are centrifuged at 3600 x g for 10 minutes. The medium is carefully aspirated and the blocks are frozen at -80°C or -20°C for 10 minutes. The blocks are then allowed to thaw at room temperature and 1 mL lysis buffer (100 mL Bugbuster supplemented with 200 ⁇ L lysonase (EMD chemicals, cat. no 71370) and protease inhibitors "complete mini EDTA-free" (Roche, cat. no. 11 836 170 001)) is added into each well.
- lysis buffer 100 mL Bugbuster supplemented with 200 ⁇ L lysonase (EMD chemicals, cat. no 71370) and protease inhibitors "complete mini EDTA-free" (Roche, cat. no. 11 836 170 001)
- the pellets are resuspended by repeat pipetting until no clump is visible and transferred to eppendorf tubes, followed by a 10-20 minute incubation on a shaker at room temperature. After centrifugation at 16,000 g for 10 minutes at 4°C, the lysates are loaded onto a TurboFilter 96 Plate included in the Ni-NTA Superflow 96 BioRobot Kit (Qiagen, cat. no. 969261) and centrifuged at 500 g for 5-10 minutes.
- the stationary phase culture is transferred into 500-mL bottles and centrifuged at 6,000 g for 10 minutes.
- the medium is decanted and the pellet is stored at -80°C or -20°C before further processing.
- the pellet is then allowed to thaw at room temperature and 20 mL lysis buffer is added into each bottle.
- the pellets are resuspended by repeat pipetting until no clump is visible, followed by 20 minute incubation on a shaker at room temperature. After centrifugation at 10,000 g for 30 minutes at 4°C, the lysates are transferred to clean tubes or bottles. If trace amounts of debris are carried over during the transfer, the sample is centrifuged again or passed through a 0.45 ⁇ m cellulose acetate membrane (Corning, cat. no. 430314) for further clarification.
- Affinity Purification A QIAFilter 96 Plate is loaded with 200 ⁇ L Ni-NTA Superflow slurry included in the Ni-NTA Superflow 96 BioRobot Kit and the resin is equilibrated by adding 600 ⁇ L binding buffer (20 mM sodium phosphate, 500 mM sodium chloride and 10 mM imidazole, pH 7.5). A vacuum of -15 in. Hg is applied until all the buffer has passed through the resin. The clarified cell lysates from the previous step are then loaded onto the QIAFilter® 96 Plate and allowed to bind for 5 minutes. A vacuum of -3 in. Hg is applied for approximately 5 minutes until all the samples have passed through the resin.
- the resin is then washed with 1 mL binding buffer, followed by two washes with 1 mL binding buffer containing 0.1% Triton X-100. The resin is then washed 10 times with 1 mL binding buffer without Triton X-100.
- the bound 6xHis-tagged ArgRS polypeptides are eluted with 450 ⁇ L elution buffer (20 mM sodium phosphate, 500 mM sodium chloride and 500 mM imidazole, pH 7.5) and stored at 4°C.
- an empty disposable column "Poly-Prep" Bio-Rad, cat. no. 731-1550
- 1 mL Ni-NTA Superflow slurry Qiagen, cat. no. 30450
- the clarified cell lysate from the previous step is then loaded onto the column and allowed to pass through by gravity.
- the resin is first washed with 50 mL binding buffer plus 0.1% Triton X-100, then washed with 50 mL binding buffer without Triton X-100.
- the bound 6xHis-tagged ArgRS polypeptides are eluted with 2 mL elution buffer and stored at 4°C.
- ArgRS polypeptides are recovered by adding 180 ⁇ L of 1X PBS pH 7.4 to each well, pipetting up and down 10 times carefully and then transferred to a clean block. This step is repeated to yield a total volume of 360 ⁇ L per well and the block is stored at 4°C.
- the eluates from Ni-NTA are loaded onto an Amicon Ultra-15 Centrifugal Filter Unit with Ultracel-3 membrane (Millipore, cat. no. UFC900308), followed by the addition of 10 mL 1X PBS and a centrifugation at 3,600 g for 10-30 minutes until the volume is less than 360 ⁇ L.
- the samples are recovered and 1X PBS is added to a final volume of 360 ⁇ L.
- an AcroPrep Advance filter plate with Mustang Q membrane (Pall, cat. no. 8171) is rinsed with 300 ⁇ L of 1X PBS and centrifuged at 1,000 g for 5 minutes to remove the buffer.
- the desalted ArgRS polypeptides (360 ⁇ L/well) are added to the filter plate and incubated on a shaker for 5-10 minutes. The plate is then centrifuged at 1,000 g for 5-10 minutes and the flow through fractions containing the ArgRS polypeptides are collected and stored at 4°C.
- the eluates from Ni-NTA are loaded onto an Amicon Ultra-15 Centrifugal Filter Unit with Ultracel-3 or Ultracel-10 membrane (Millipore, cat. no. UFC900308 or UFC901008) depending on the molecular weight of the ArgRS polypeptide and then centrifuged at 3,600 g for 10-30 minutes until the volume is reduced to 250 ⁇ L.
- the samples are mixed in 10 mL 1X PBS, pH7.4 and centrifuged again at 3,600 g for 10-30 minutes until the volume is about 250 ⁇ L. This step is repeated one more time, the supernatants are recovered and 1X PBS is added to a final volume of 1.5 mL.
- a Sartobind Q 5 strong anion exchanger membrane (Sartorius, cat. no. Q5F) is flushed with 1 mL 1X PBS and the ArgRS polypeptides are slowly passed through the membrane using a plastic syringe. The flow through fraction containing the ArgRS polypeptides is collected in a 96-deep well block that is sealed and stored at 4°C.
- 6xHis-tagged ArgRS polypeptides expressed in bacteria and found in inclusion bodies are purified using affinity chromatography and a series of refolding steps, as described below.
- Bacterial cuftures 100 ng of plasmid encoding each ArgRS polypeptide is transformed into BL21(DE3) (EMD chemicals, cat. no. 69450) or C41(DE3) (Lucigen, cat. no. 60442) competent E. coli bacteria at 42°C for 30 seconds in PCR plates. The plates are placed on ice for 2 minutes and 100 ⁇ L of SOC medium is added, followed by a 1-hour incubation at 37°C. 5 mL of auto-induction medium (EMD chemicals, cat. no. 71491) supplemented with kanamycin (100 ⁇ g/mL) is added into each well of a 24-well block (Qiagen, cat. no. 19583). The transformation reactions are added to the individual wells, the block is sealed with adhesive film (VWR, cat. no 60941-078) and incubated overnight at 250 rpm in a 37°C shaker.
- VWR adhesive film
- pellets are resuspended by repeat pipetting until no clump is visible and transferred to eppendorf tubes, followed by a 10-20 minute incubation on a shaker at room temperature. After centrifugation at 16,000 x g for 10 minutes at 4°C, the soluble lysates are discarded and the inclusion bodies are thoroughly resuspended in denaturing binding buffer (20 mM sodium phosphate, 500 mM sodium chloride, 6 M guanidine hydrochloride, 10 mM imidazole, pH 7.5).
- denaturing binding buffer (20 mM sodium phosphate, 500 mM sodium chloride, 6 M guanidine hydrochloride, 10 mM imidazole, pH 7.5).
- the samples are centrifuged at 16,000 g for 10 minutes and the supernatants loaded onto a TurboFilter 96 Plate included in the Ni-NTA Superflow 96 BioRobot Kit (Qiagen, cat. no. 969261) followed by centrifugation at 500 g for 5-10 minutes.
- the filtrates are collected in a clean 96-well block (Greiner, cat. no. 780286).
- the stationary phase culture is transferred into 500-mL bottles and centrifuged at 6,000 g for 10 minutes.
- the medium is decanted and the pellet is stored at -80°C or -20°C before further processing.
- the pellet is then allowed to thaw at room temperature and 20 mL lysis buffer is added into each bottle.
- the pellets are resuspended by repeat pipetting until no clump is visible, followed by 20 minute incubation on a shaker at room temperature. After centrifugation at 10,000 g for 30 minutes at 4°C, the soluble lysates are discarded and the insoluble inclusion bodies thoroughly resuspended in denaturing binding buffer.
- Affinity Purification A QIAFilter 96 Plate is loaded with 200 ⁇ L Ni-NTA Superflow slurry included in the Ni-NTA Superflow 96 BioRobot Kit and the resin is equilibrated by adding 600 ⁇ L denaturing binding buffer (see above). A vacuum of -15 in. Hg is applied until all of the buffer passes through the resin. The clarified denatured samples from the previous step are then loaded onto the QIAFilter® 96 Plate and allowed to bind for 5 minutes. A vacuum of approximately 3 inches of mercury is applied for approximately 5 minutes until all the samples pass through the resin.
- the resin is then washed with 1 mL denaturing binding buffer, followed by five washes with 1 mL denaturing binding buffer containing 0.1% Triton X-100. The resin is then washed 15 times with 1 mL denaturing binding buffer without Triton X-100.
- the bound 6xHis-tagged ArgRS polypeptides are then eluted with 450 ⁇ L denaturing elution buffer (20 mM sodium phosphate, 500 mM sodium chloride, 6 M guanidine hydrochloride and 500 mM imidazole, pH 7.5) and stored at 4°C.
- an empty disposable column "Poly-Prep" Bio-Rad, cat. no. 731-1550
- 1 mL Ni-NTA Superflow slurry Qiagen, cat. no. 30450
- the denatured inclusion bodies from the previous step are then loaded onto the column and allowed to pass through by gravity.
- the resin is first washed with 50 mL denaturing binding buffer plus 0.1% Triton X-100, then washed with 50 mL denaturing binding buffer without Triton X-100.
- the bound 6xHis-tagged ArgRS polypeptides are eluted with 2 mL denaturing elution buffer and stored at 4°C.
- ArgRS polypeptides are recovered by adding 200 ⁇ L of refolding buffer containing 50 mM Tris, 250 mM sodium chloride, 10 mM potassium chloride, 2 mM magnesium chloride, 2 mM calcium chloride, 400 mM sucrose, 500 mM arginine, 1 mM DTT and 0.01% polysorbate 80, pH 7.4) to each well, pipetting up and down 10 times carefully, and then transferred to a clean block. This step is repeated to yield a total volume of 400 ⁇ L per well and the block is placed on the shaker overnight at 4°C.
- ArgRS polypeptides ⁇ 10 kDa the eluates from Ni-NTA are loaded onto an Amicon Ultra-15 Centrifugal Filter Unit with Ultracel-3 membrane (Millipore, cat. no. UFC900308), followed by the addition of 10mL refolding buffer and a centrifugation at 3,600 g for 10-30 minutes until the volume is less than 400 ⁇ L. The samples are recovered and extra refolding buffer is added to a final volume of 400 ⁇ L. The samples are transferred to a 96-well block, sealed with film and placed on a shaker overnight at 4°C.
- the eluates from Ni-NTA are loaded onto an Amicon Ultra-15 centrifugal filter unit with Ultracel-3 or Ultracel-10 membrane (Millipore, cat. no. UFC900308 or UFC901008 depending on the molecular weight of the ArgRS polypeptide) and then centrifuged at 3,600 g for 10-30 minutes until the volume is reduced to about 500 ⁇ L.
- Ultracel-3 or Ultracel-10 membrane Millipore, cat. no. UFC900308 or UFC901008 depending on the molecular weight of the ArgRS polypeptide
- the samples are diluted 20-fold in the following buffer: 50 mM sodium acetate, 10 mM sodium chloride, 0.4 mM potassium chloride, 1 mM EDTA, 400 mM sucrose, 500 mM arginine, 1 mM DTT and 0.01% polysorbate 80, pH 6.0.
- the samples are diluted 20-fold in the following buffer: 50 mM Tris, 250 mM sodium chloride, 10 mM potassium chloride, 2 mM magnesium chloride, 2 mM calcium chloride, 400 mM sucrose, 500 mM arginine, 1mM DTT and 0.01% polysorbate 80, pH 8.0.
- the samples are incubated on a shaker at 4°C overnight.
- the 96-well block is centrifuged at 3,600 g to remove any potential aggregates. The supernatants are then subjected to buffer exchange with 1X PBS (Invitrogen, cat. no. 10010).
- 1X PBS Invitrogen, cat. no. 10010
- the Omega 10K membrane of an AcroPrep 96 filter plate is rinsed with 20 ⁇ L 1X PBS and the plate is placed onto a vacuum manifold (>10 in. Hg) until all the liquid passes through.
- the samples in the refolding buffer are dispensed into each well and the vacuum applied until all the liquid passes through. These steps are repeated until the total sample volume (400 ⁇ L) has been processed.
- ArgRS polypeptides are recovered by adding 180 ⁇ L of 1X PBS pH 7.4 to each well, pipetting up and down 10 times carefully, and then transferred to a clean block. This step is repeated to yield a total volume of 360 ⁇ L per well and the block is stored at 4°C.
- the refolded samples are loaded onto an Amicon Ultra-15 Centrifugal Filter Unit with Ultracel-3 membrane (Millipore, cat. no. UFC900308) followed by the addition of 10 mL 1X PBS and centrifugation at 3,600 g for 10-30 minutes until the volume is less than 360 ⁇ L. The samples are recovered and 1X PBS is added to a final volume of 360 ⁇ L.
- an AcroPrep Advance filter plate with Mustang Q membrane (Pall, cat. no. 8171) is rinsed with 300 ⁇ L of 1X PBS and centrifuged at 1,000 g for 5 minutes to remove the buffer.
- the ArgRS polypeptides (360 ⁇ L/well) are added to the filter plate and incubated on a shaker for 5-10 minutes. The plate is then centrifuged at 1,000 g for 5-10 minutes and the flow through fractions containing the ArgRS polypeptides are collected and stored at 4°C.
- the refolded samples are centrifuged at 10,000 g for 10 minutes to remove any insoluble aggregates.
- the supernatant is loaded onto an Amicon Ultra-15 Centrifugal Filter Unit and centrifuged at 3,600 g until the volume is reduced to 250 ⁇ L.
- the samples are mixed in 10 mL 1X PBS and centrifuged again at 3,600 g for 10-30 minutes until the volume is about 250 ⁇ L.
- the pH of 1X PBS is adjusted to match the pH of the refolding buffer, either pH 6.0 or pH 8.0. This step is repeated one more time, the supernatants are recovered and 1X PBS is added to a final volume of 1.5 mL.
- a Sartobind Q 5 strong anion exchanger membrane (Sartorius, cat. no. Q5F) is flushed with 1 mL 1X PBS and the ArgRS polypeptides are slowly passed through the membrane using a plastic syringe. The flow through fraction containing the ArgRS polypeptides is collected in a 96-deep well block that is sealed and stored at 4°C.
- BIOPHYSICAL CHARACTERIZATION All purified ArgRS polypeptides are analyzed by SDS-PAGE, their concentration determined based on A 280 and calculated extinction coefficient (ProtParam on ExPASy server). Endotoxin levels are measured by the QCL-1000 Endpoint Chromogenic LAL assay (Lonza, cat. no. 50-648U) according to the manufacturer's instructions.
- Dynamic Light Scattering A Wyatt Technology DynaPro 99 instrument and the temperature controller (20°C) are warmed up for 15 minutes before the experiment followed by connection of the Dynamics software to the instrument. The acquisition time is set to 10 seconds for multiple acquisitions and the laser power is set to 100%.
- the quartz cuvette is washed thoroughly with deionized water and methanol before the addition of the protein sample (15 ⁇ L at a concentration of approximately 1 mg/mL in PBS). Air bubbles are removed by tapping the cuvette before it is inserted into the holder with the frosted side to the left. If the intensity is too high (warning message shown on the screen), the sample is further diluted with PBS until the intensity is decreased to a normal range.
- the data collected include hydrodynamic radius, polydispersity, predicted average molecular weight, percentage of intensity and percentage of mass.
- the protein sample is diluted to a concentration of about 5-10 mg/mL in PBS before being loaded into a 100 ⁇ L sample loop on the General Electric AKTA FPLC.
- the Superdex 200 10/300 GL size exclusion column (General Electric, cat. no. 17-5175-01) is used for separation.
- the column is first equilibrated with 1.5 column volume (CV) of 1X PBS buffer, followed by sample injection.
- the column is run in 1 CV of 1x PBS buffer (isocratic flow) with absorbance at 280nm monitoring.
- the peak area is integrated and the percentage calculated with the Unicorn software.
- the elution volume is used to estimate the molecular weight based on comparison with gel filtration calibration kits (General Electric, cat. no. 28-4038-41 and 28-4038-42).
- ArgRS polypeptides Purified ArgRS polypeptides (1 mg/mL) are diluted 1:10 into 0.1% formic acid and 0.6 ⁇ g protein is loaded with a Dionex autosampler onto a C4 capillary column.
- the capillary column is prepared by cutting 150 mm of fused silica tubing (0.36 mm OD by 0.1 mm ID, Polymicro Technologies, cat. no. 2000023). The capillary is pulled at one end with a Suter Instrument Laser Fiber Puller and cut with a fused silica cutter to generate a 5 ⁇ m tip.
- the capillary is packed to the length of 75 mm with C4 resin (5 ⁇ m, 300 ⁇ , Michrom, cat. no.
- the LC-MS analysis is performed on an ThermoFisher LTQ ion trap mass spectrometer coupled to a Dionex Ultimate3000 HPLC system.
- the analyte is eluted from the column using a 35-minute gradient of 5-70% acetonitrile in 0.1% formic acid at a flow rate of 0.9 ⁇ L/min.
- the LTQ is operated on a full MS scan mode (300-2,000 m/z) with a spray voltage of 2.5 kV.
- raw mass spectrometry data are stored in RAW files generated by XCalibur running on the LTQ XL mass spectrometer.
- MS spectra of the major peaks on the chromatograph are further analyzed with ThermoFisher deconvoluting algorithm ProMass to obtain the ArgRS polypeptide molecular weights.
- the cell types chosen for transcriptional profiling are based on the pluripotent capabilities of the cells in question and potential to identify ArgRS polypeptides of direct therapeutic value.
- MSCs Mesenchymal stem cells
- marrow stromal cells can also be induced to differentiate into cells of different connective tissue lineage, such as bone, cartilage, and fat.
- connective tissue lineage such as bone, cartilage, and fat.
- hMSCs Human Mesenchymal stem cells
- HMSCs are capable of cell fate crossing germ layer boundaries.
- these cells can also differentiate into neurons of ectodermal origin and hepatocyte-like cells of endodermal origin. During the process of differentiation, these cells may modify expression patterns of certain lineage specific transcripts.
- ArgRS polypeptides to modulate specific patterns of genes in HMSCs in a time dependent manner demonstrates that these proteins play potentially significant roles in a broad array of differentiation pathways, as well as diseases and disorders resulting from the dysfunction, or deterioration of these processes, or the corresponding cell types.
- ArgRS polypeptides with the ability to modulate gene transcription in MSCs have significant therapeutic utility to enable the in vitro or in vivo modulation of hematopoiesis, neurogenesis, myogenesis, osteogenesis, and adipogenesis, as well as in a broad range of disorders and diseases, including for example inflammatory responses, autoimmunity, cancer, neuronal degeneration, muscular dystrophy, osteoporosis, and lipodystrophy.
- Human Skeletal Muscle Cells can undergo differentiation to exhibit actin and myosin myofilaments, and have been used in the study of genetic muscular diseases such as Malignant Hyperthermial. HSkMC also have the potential to act as a cardiac graft, mending damage to the heart. Recently, cultured Human Skeletal Muscle cells have been used in micro gravity experiments to study the effects of low gravity environments on Human Skeletal Muscle.
- ArgRS polypeptides Accordingly the ability of specific ArgRS polypeptides to modulate specific patterns of genes in HSkMC in a time dependent manner demonstrates that these proteins play potentially significant roles in the processes of myogenesis, as well as diseases and disorders resulting from the dysfunction, or deterioration of these processes as well as muscle cell development or metabolism. Accordingly ArgRS polypeptides with the ability to modulate gene transcription in muscle cells have therapeutic utility in a broad range of diseases including for example, the treatment of metabolic disease, cachexia, various muscle wasting conditions, as well as musculoskeletal diseases.
- HMSC Human Marrow Stromal Cells
- HSkMC Human Skeletal Muscle Cells
- Controls include Differentiation media with a standard cocktail to promote (1) Adipogenesis, (2) Osteogenesis, (3) Chondrogenesis and (4) Skeletal muscle myotube formation. Additional controls include untreated wells containing only growth media. Two wells were run for each Differentiation control. Controls: all media was made utilizing DMEM as the basal media. Standard literature was followed and Differentiation media was purchased from Cell Applications.
- differentiation media contained the following additives: Skeletal muscle differentiation cocktail: FBS, insulin, glutamine, FGF, EGF; Adipogenesis cocktail: insulin, dexamethasone and IBMX; Osteogenesis cocktail: FBS, dexamethasone, ascorbate 2 phosphate, beta-glycerophosphate; Chondrogenesis cocktail: insulin, ascorbate-2-phosphate, and TGF- ⁇ 1.
- Skeletal muscle differentiation cocktail FBS, insulin, glutamine, FGF, EGF
- Adipogenesis cocktail insulin, dexamethasone and IBMX
- Osteogenesis cocktail FBS, dexamethasone, ascorbate 2 phosphate, beta-glycerophosphate
- Chondrogenesis cocktail insulin, ascorbate-2-phosphate, and TGF- ⁇ 1.
- Standard protocols for using an ABI (Applied Biosystems, Item # AM1728) TAQMAN® Gene Expression Cells-to-CTTM Kit are utilized to lyse cells and harvest genomic material.
- An ABI Pre-Amp Mix (Applied Biosystems, Item# 4391128) is used to initiate pre-amplification.
- Gene specific primers are created using a Primer 3 program and purchased from IDT technologies.
- Fluidigm profiling arrays (Item # BMK-M-96.96) were used for actual quantitative PCR with standard Fluidigm loading reagents and pipetting devices. Table E1 below lists the genes profiled.
- Bioinformatics Analysis Data retrieved in .csv format from the Biomark machine by Fluidigm is converted to a tabular format including sample, mRNA, and replicate information along with the raw fluorescence value. PCR reactions that failed are marked as missing. Multiple experiments were combined after normalizing to total expression of mRNA species. All measured mRNA expression is filtered based on the requirement of detection in at least 2 of all of the biological replicates tested. We assessed technical, biological and set deviation mean in entire dataset.
- up-regulated genes i.e. ⁇ Cts greater than 0
- Fold Change 2 ⁇ Ct.
- down-regulated genes i.e. ⁇ Cts less than 0
- Fold Change -(2 ⁇
- ArgRS polypeptides with the ability to modulate the rate of cellular proliferation and or apoptosis have significant therapeutic utility in a broad range of diseases including, as growth factors, and differentiation factors for stem cells, and in treatment regimens to enhance or suppress the proliferation of specific cell types of interest in vivo or in vitro, including for example, haemopoietic cells, immunomodulatory cells, cancer, and for the treatment and prevention of diseases associated with aging, including for example neurodegeneration, peripheral neuropathy, and loss of muscular and soft tissue tone.
- Hoechst 33432 Standard cell counts to assess proliferation are performed using Hoechst 33432, which is a cell-permeant nuclear counterstain that emits blue fluorescence when bound to dsDNA. It is available as a solution (Invitrogen Cat # H-3570) that is used at a final concentration of 1ug/mL in either media or PBS. Cells are grown in 96 well plates in the presence of ArgRS polypeptides for a standard growth time of 48 hours, or longer depending on cell type and as described in the examples below.
- ATP-lite Cellular ATP levels correlate with cellular health and can be readily determined using a variety of commercially available kits.
- ATP-lite Perkin-Elmer, Cat #6016947 Boston, MA 02481 which is a homogenous mixture of lysis solution and ATP-detection reagent. is pre-mixed before use and is used 1:1 v:v ratio with cultured cells. Plates are incubated for 5 minutes to promote lysis and plates are measured using a luminescent plate reader. Cells are grown in 96 well plates in the presence of ArgRS polypeptides for a standard growth time of 48 hours, or longer depending on cell type and as described in the examples below.
- ALAMARBLUE ® is a cell viability indicator which is based on the redox state of the cells.
- Resazurin the active ingredient, is a nontoxic, cell permeable compound that is blue in color and virtually nonfluorescent when present in its oxidized form.
- resazurin is rapidly reduced to resorufin, which produces a red fluorescence signal.
- Viable cells continuously convert resazurin to resorufin, thereby generating a quantitative measure of viability-and cytotoxicity. The lack of toxicity allows long-term exposure of cells to resazurin without negative impact; cells grown in the presence of resazurin were found to produce similar numbers of viable cells as control cells, as determined by flow cytometric analysis.
- Measurements are made by adding a solution of Resazurin / ALAMARBLUE® to cells, incubating them for 1-4 hours, and reading the fluorescence or absorbance.
- the amount of fluorescence or absorbance is proportional to the number of living cells and corresponds to the cells metabolic activity. Damaged and nonviable cells have lower innate metabolic activity and thus generate a proportionally lower signal than healthy cells.
- samples can readily be measured on fluorescence and absorbance instrumentation. For fluorescence readings: 530 nm excitation and 590 nm emission filter settings are used.
- Cells are grown in 96 well plates in the presence of ArgRS polypeptides for a standard growth time of 48 hours, or longer depending on cell type and as described in the examples below.
- LDL is the major carrier of cholesterol in the blood, accounting for more than 60% of the cholesterol in plasma.
- the hepatic LDL receptor is responsible for clearing around 70 % of plasma LDL from circulation. Internalized LDL is degraded to free cholesterol and amino acids in the lysosome.
- the liver is the most important organ for LDL catabolism and LDL receptor activity in humans. LDL that is not internalized and remains in circulation can be transported by endothelial cells into the vessel wall, resulting in the formation of atherosclerotic plaques. Circulating LDL can also be taken up by macrophages and this can also contribute to the formation of plaques.
- ArgRS polypeptides can regulate uptake of acetylated LDL
- a standard assay for measuring acetylated LDL uptake is employed in HepG2C3a cells.
- ArgRS polypeptides with the ability to modulate LDL uptake have significant therapeutic utility in a broad range of diseases including for example, the treatment of hypercholesteremia, hyperlipidemia, type 1 and 2 diabetes, metabolic syndrome, and vascular diseases including atherosclerosis
- HEPG2C3a cells (ATCC# CRL-10741) are maintained in Eagles Minimal Essential (EMEM) medium supplemented with 10% FBS (HyClone Cat#SH30910.03), 50u/mL penicillin/50 ⁇ g/mL streptomycin, (Invitrogen) in 15 mL medium in 75 mL flasks. Cells are grown at 37°C, 5% CO 2 , in a humidified environment and utilized in BSL2 certified tissue culture hoods using sterile technique and appropriate personal protective equipment including goggles, gloves and lab coats.
- HEPG2C3a express the LDL-receptor and are competent for acetylated LDL uptake when grown on clear bottom collagen coated plates.
- a 100 ⁇ L volume of cells is plated on collagen coated plates (Invitrogen Cat#A11428) overnight in complete medium (above) at a cell density of 50,000 cells/mL.
- Cells are washed once with PBS (Invitrogen Cat# 10010) and 80 ⁇ L of serum free EMEM is added to each well.
- ArgRS polypeptides at a final concentration of 250nM per well are added in a consistent volume in sterile PBS to each well.
- a unique ArgRS polypeptide is placed in each well. Cells are serum starved and exposed to the ArgRS polypeptides for 16 hours.
- soluble ICAM is measured using a standard ELISA kit from RND Systems (Cat # DY643), and serum free media supplemented with 5 ⁇ g/mL ac-LDL (Alexa Fluor 488 labeled Cat # L23380, Invitrogen) is added to each well.
- ac-LDL Alexa Fluor 488 labeled Cat # L23380, Invitrogen
- Plates were analyzed for total fluorescent intensity using a bottom read on a Victor X5 fluorescent plate reader (Perkin Elmer) at an excitation wavelength centered around 485 nm, and an emission wavelength centered around 535 nm. Cells are stained with Hoechst dye and fluorescent intensity 405nm Excitation / 450nM Emission is read to confirm total cell number is consistent across the plate.
- Phagocytosis by polymorphonuclear neutrophils and monocytes constitutes an essential arm of host defense against infections by microorganisms including bacteria and fungi.
- the phagocytic process can be separated into several major stages: chemotaxis (migration of phagocytes to inflammatory sites), attachment of particles to the cell surface of phagocytes, ingestion (phagocytosis) and intracellular killing by oxygen-dependent (oxidative burst) and oxygen-independent mechanisms.
- chemotaxis miration of phagocytes to inflammatory sites
- phagocytosis ingestion
- oxygen-dependent burst oxygen-dependent burst
- Reduced or missing burst activity is observed in inborne defects like the chronic granulomatous disease (CGD).
- CGD chronic granulomatous disease
- the disease is characterized by repeated and life-threatening infections caused by bacterial and fungal organisms. These infections typically consist of pneumonia, lymphadenitis, or abscesses that involve lymph nodes, lungs, and liver.
- the NADPH oxidase is the enzyme system responsible for producing superoxide anion, which is quickly converted to hydrogen peroxide and hydroxyl radicals. Abnormalities in the constituent peptides of the NADPH oxidase enzyme system lead to the dysfunctions characteristic of CGD. Neutrophils from CGD patients fail to produce a significant oxidative burst following stimulation. Different forms of CGD are described (classical X-linked CGD and autosomal recessive patterns).
- oxidative burst of granulocytes is impaired in transplantation, later stages of HIV infection, and in the elderly, making these populations more susceptible to secondary infection and exacerbations of inflammatory disease.
- Various immunomodulators e.g., cytokines (GM-CSF, G-CSF, TNF) or drugs
- proteins with the ability to up-regulate or down-regulate oxidative burst in a therapeutic fashion to be useful for a variety of different disease states.
- the protein kinase C ligand phorbol 12-myristate 13-acetate (PMA) can be utilized in this assay as an agonist of the oxidative burst process. Heparinized whole blood is mixed with sterile dextran (0.6% final concentration) for 1 hour and allowed to separate into layers. The lower layer contains neutrophil, monocytes and red blood cells. An ammonium chloride lysis step is utilized to remove all RBCs and a 97% pure population of neutrophils with approximately 3% monocyte contamination remains following lysis step. Upon stimulation, granulocytes and monocytes produce reactive oxygen metabolites (superoxide anion, hydrogen peroxide, hypochlorous acid) which destroy bacteria inside the phagosome.
- reactive oxygen metabolites superoxide anion, hydrogen peroxide, hypochlorous acid
- Formation of the reactive oxidants during the oxidative burst can be monitored by the addition and oxidation of Amplex Red. The percentage of cells having produced reactive oxygen radicals are then analyzed as well as their mean fluorescence intensity using a fluorescent plate reader. The typical time course for this reaction is 10 minutes, with obvious burst being seen by 2 minutes and a drop off of signal being seen by 20 minutes.
- This assay can be run in agonist mode in the absence of PMA or in antagonist mode, with concomitant administration of ArgRS polypeptides and PMA at a concentration that is below the EC50 for this compound.
- Neutrophil elastase is a serine protease that has been implicated as having a specific role in the development of a wide range of human diseases, including inflammatory disorders of the lung and cardiovascular system. Although its key physiologic role is in innate host defense, it can also participate in tissue remodeling and possesses secretagogue actions that are now recognized as important to local inflammatory signals. Neutrophil elastase activity has been implicated in the development of emphysema for several decades, however only relatively recently has a pathogenetic function been ascribed to this serine proteinase in situations where excessive extracellular matrix deposition occurs. The use of genetically manipulated animal models is starting to uncover the potential ways in which its actions might influence fibrotic lung repair.
- This assay uses the ENZCHEK® Elastase Assay Kit (Invitrogen Catalog # E-12056). Neutrophils are prepared from fresh human blood using a 6% dextran solution and red blood cells are lysed before plating cells in RPMI media (media should be un-supplemented with no serum, no antibiotics). A 1.0 mg/mL stock solution of the DQ elastin substrate is prepared by adding 1.0 mL of deionized water (dH2O) directly to one of the three vials containing the lyophilized substrate and mixing to dissolve. 1X Reaction Buffer is prepared by diluting 6 mL of the 10X Reaction Buffer in 54 mL dH2O.
- dH2O deionized water
- a 100 ⁇ g/mL working solution of the DQ elastin substrate is prepared by diluting the DQ elastin stock solution tenfold in 1X Reaction Buffer.
- Porcine pancreatic elastase stock solution is prepared by making a 100 U/mL stock solution in dH2O.
- 50 ⁇ L of 1X Reaction Buffer is pipette into each assay well containing 500,000 neutrophils/ mL in a 30 ⁇ L volume. 8 ⁇ L of each ArgRS polypeptide is added per well, and the sample incubated for 20 minutes at 37 °C.
- 50 ⁇ L of 100 ⁇ g/mL DQ elastin working solution is added to each well and mixed. Samples are incubated at room temperature, protected from light, for 30 minutes. Fluorescence intensity in a fluorescence microplate reader equipped with standard fluorescein filters (ex 485/ Em 535) fluorescence may be measured over multiple time points.
- Macrophages are major players in the innate immune system and express a large repertoire of different classes of pattern recognition receptors (PRRs), including the family of Toll-like receptors (TLRs) which are powerful regulators and controllers of the immune response.
- PRRs pattern recognition receptors
- TLRs Toll-like receptors
- TLRs Stimulation of TLRs by microbial pathogens and endogenous ligands initiates signaling cascades that induce the secretion of pro-inflammatory cytokines and effector cytokines that direct downstream adaptive immune responses.
- Endogenous ligands, as well as microbial components, are recognized by and can activate TLRs, raising the possibility that these receptors may be critical targets for the development of new therapies for multiple diseases.
- ArgRS polypeptides that modulate TLR receptor activity have therapeutic utility in a broad range of diseases and disorders including for example, inflammatory diseases and disorders, autoimmune diseases, tissue transplantation / organ rejection, cancer prevention or treatment, the modulation of haematopoiesis and infection.
- Mouse macrophages sold under the trademark RAW-BLUETM cells express all TLRs except TLR5 and include a secreted embryonic alkaline phosphatase (SEAP) gene which is inducible by NF-kB and AP-1 transcription factors.
- SEAP embryonic alkaline phosphatase
- RAW-BLUETM cells activate NF-kB and/or AP-1 leading to the secretion of SEAP which is measurable when using SEAP detection medium.
- RAW-BLUETM cells are washed twice with PBS, trypsinized and resuspended in fresh Growth Medium (Growth Medium: DMEM, 4.5 g/l glucose, 10% heat-inactivated fetal bovine serum (30 minutes at 56°C), 100 mg/mL ZEOCINTM, 2 mM L-glutamine).
- DMEM fresh Growth Medium
- ZEOCINTM 100 mg/mL ZEOCINTM
- Cells are plated at a concentration of 50,000 cells/well in a 96 well plate in a total volume of 100 ⁇ L, and ArgRS polypeptides, controls, or ArgRS polypeptides (+LPS) are added to each well at the concentrations shown in the experiments outlined below. Cells are incubated at 37°C in a 5% CO 2 incubator for 18 hours.
- SEAP detection medium (QUANTI-BLUETM) (Invivogen Catalog code: rep-qbl) is prepared following the instructions and 120 ⁇ L is added per well to a clear flat-bottom 96-well plate, and cell supernatant is added (20 ⁇ L). Samples are incubated at 37°C for about 30 minutes to up to 2 hours. SEAP levels are determined using a spectrophotometer and reading absorbance at 650 nM.
- this assay can be modified to identify potential TLR antagonists.
- ArgRS polypeptides are added to the cells at a final concentration of about 250nM per well, (or as otherwise specified in the Examples below) 1 hour prior to adding 50 ng/mL LPS. Cells are incubated and SEAP detected as described above. PBS control wells with no LPS or ArgRS polypeptide alone added are used to find the basal level of TLR stimulation at the time of the measurement. Control wells are pretreated with PBS and known TLR agonists and antagonists. The ratio of the background subtracted [PBS plus LPS signal] to [ArgRS polypeptide plus LPS signal] is used to determine percent antagonism.
- Human HEK293 cells are genetically modified and sold under the trademark HEK-BlueTM TLR cells (Invivogen).
- the TLR2 and TLR4 versions of this cell type selectively express all TLR2 or TLR4 and include a secreted embryonic alkaline phosphatase (SEAP)reporter gene under the control of an IFN-beta minimal promoter which is fused to five NF-kB and AP-1 transcription factors binding sites.
- SEAP embryonic alkaline phosphatase
- HEK-BLUETM TLR2 and HEK-BLUETM TLR4 cells activate NF-kB and/or AP-1 leading to the secretion of SEAP which is measurable when using SEAP detection reagent.
- the HEK-BLUETM TLR2 cells are co-transfected with the LPS co-receptor protein CD 14 to enhance TLR2 responsiveness and improve signal quality.
- the parent cell expresses endogenous levels of TLRl, 3, 5, 6 and also NOD1.
- HEK-BLUETM -TLR2 or HEK-BLUETM -TLR4 cells are washed twice with PBS, trypsinized and resuspended in fresh Growth Medium (Growth Medium: DMEM, 4.5 g/L glucose, 10% heat-inactivated fetal bovine serum (30 minutes at 56°C), 100 mg/mL ZEOCINTM, 2 mM L-glutamine).
- Growth Medium DMEM, 4.5 g/L glucose, 10% heat-inactivated fetal bovine serum (30 minutes at 56°C), 100 mg/mL ZEOCINTM, 2 mM L-glutamine.
- Cells are plated at a concentration of 50,000 cells/well in a 96 well plate in a total volume of 100 ⁇ L, and ArgRS polypeptides, controls, or ArgRS polypeptides (+LPS) are added to each well at the concentrations shown in the experiments outlined below.
- SEAP detection medium (QUANTI-BLUETM) (Invivogen Catalog code: rep-qbl) is prepared following the instructions and 120 ⁇ L is added per well to a clear flat-bottom 96-well plate, and cell supernatant is added (20 ⁇ L). Samples are incubated at 37°C for about 30 minutes to up to 2 hours. SEAP levels are determined using a spectrophotometer and reading absorbance at 650 nM. Control wells are pretreated with PBS and known TLR agonists such as UltraPure LPS (TLR-4) or PAM3CSK4 (TLR-2). The ratio of the background subtracted [PBS plus LPS signal] to [ArgRS polypeptide plus LPS signal] is used to determine percent agonism.
- TLR-4 UltraPure LPS
- TLR-2 PAM3CSK4
- Cytokines are a diverse set of small cell signaling protein molecules that are used extensively for intercellular communication, and play significant roles in normal body homeostasis, including immunomodulation and regulation. Accordingly ArgRS polypeptides that modulate the release, or biological activities of cytokines, have therapeutic utility in a broad range of diseases and disorders including for example, inflammatory diseases and disorders, autoimmune diseases, tissue transplantation / organ rejection, cancer prevention or treatment, the modulation of haematopoiesis and infection.
- Test cells are seeded into a 24-well plate at density of about 1 million cells/well in 1 mL of growth media. Cells are treated with either ArgRS polypeptide (at the concentrations shown in the examples below) or an equal volume of PBS and incubated overnight at 37° with 5% CO 2 . Following cell treatment, samples are centrifuged at 4 °C in a swinging bucket centrifuge at 2,000 x g for 5 minutes. Media is carefully removed so as to not disturb the cell pellet and transferred to a new tube. Samples are assayed immediately or snap frozen in liquid nitrogen for subsequent analysis.
- Cytokine release (including the cytokines MIF, IL-8, IL-10, Serpin E1, GM-CSF, GRO, IL-1 alpha, IL-1beta, IL-1ra, IL-6, MCP-1, MIP-1, RANTES and TNFalpha) is determined using commercially available kits (R&D Systems, Inc, MN, USA) or via a contract research organization (MD Biosciences (St. Paul, MN).
- Human whole blood is obtained from normal human donors and collected with heparin in standard collection tubes. Blood is used on the same day as it is collected to ensure adequate cell health. Blood is mixed gently and plated in an 100 ⁇ L volume into 96 well polycarbonate V bottom plates. ArgRS polypeptides are added and slowly mixed into blood 2X using a multichannel pipet set on 50 ⁇ L. Filter tips are used for all experimentation and full PPE is worn. All experimentation occurs in a dedicated biosafety hood that is suitable for experimentation with human blood. Blood is incubated overnight at 37° C with 5% CO 2 . Following cell treatment, samples are centrifuged in a swinging bucket centrifuge at 2,000 x g for 5 minutes. Supernatant is collected for cytokine ELISAs ELISA are performed as described previously.
- peripheral blood mononuclear cells To isolate peripheral blood mononuclear cells freshly isolated human whole blood is gently layered over Sigma HISTOPAQUE®-1077 at a ratio of 1:1 in 50 mL conical tubes at room temperature. Layered samples are centrifuged at 400 x g in a swinging bucket clinical centrifuge for 30 minutes at room temperature with no brake. The white cellular layer at the interface between the plasma and density gradient is then removed by pipet. These peripheral blood mononuclear cells are washed twice with RPMI-1640 (Invitrogen #22400-105) by dilution and centrifugation for 10 minutes at 250 x g. The washed PBMC were resuspended in RPMI-1640 + 10% FBS and plated at 1x10 6 cells/mL.
- RPMI-1640 Invitrogen #22400-105
- IL-6 and IL-8 are overproduced in several diseases, and thus may play a fundamental role in the pathogenesis of inflammatory disease.
- IL-6 activates endothelial cell production, leading to the release of IL-8 and monocyte chemoattractant protein, expression of adhesion molecules, and recruitment of leukocytes to inflammatory sites.
- monocyte chemoattractant protein a protein that influences the production of IL-8 and monocyte chemoattractant protein
- adhesion molecules e.g., IL-8
- cytokines are expressed in cell types associated with inflammatory disease, including cells involved in the pathogenesis of systemic juvenile arthritis, systemic lupus erythematosus, Crohn's disease, and rheumatoid arthritis.
- One of the most important systemic actions of cytokine production is the induction of the acute phase response.
- Acute phase proteins are produced primarily by the liver and include proteins that promote the immune response through activation of complement, induction of proinflammatory cytokines, and stimulation of neutrophil chemotaxis.
- the acute phase response can be helpful, and acute-phase proteins, such as proteinase antagonists, opsonins, and procoagulants, help limit tissue destruction by resolving inflammation.
- IL-6 can stimulate synoviocyte proliferation and osteoclast activation, leading to synovial pannus formation and repair.
- IL-6 acts with IL-1 to increase production of matrix metalloproteinases, which may contribute to joint and cartilage destruction.
- IL-6 may also have protective effects in the joint, as suggested by the finding that this cytokine induces the expression of the tissue inhibitor of metalloproteinase and stimulates proteoglycan synthesis when injected into the joints of mice with antigen-induced arthritis.
- Human Fibroblast-Like Synoviocytes-Rheumatoid Arthritis (HFLS-RA) are isolated from synovial tissues obtained from patients with Rheumatoid Arthritis (RA). They are cryopreserved at second passage and can be cultured and propagated at least 5 population doublings.
- HFLS are long known for their role in joint destruction by producing cytokines and metalloproteinases that contribute to cartilage degradation.
- ArgRS polypeptides with the ability to modulate the growth, differentiation, or cytokine release profile of fibroblast-like synoviocytes-rheumatoid arthritis (HFLS-RA) have therapeutic utility in a broad range of diseases including for example, the treatment of inflammatory diseases and disorders including systemic juvenile arthritis, systemic lupus erythematosus, Crohn's disease, and rheumatoid arthritis.
- HFLS-RA adult cells
- Cell Applications Cat # 408RA-05a are maintained in Synoviocyte Growth Medium (Cell Applications Cat #415-50) in 15 mL medium in 125 mL flasks for 1 passage before use.
- Cells are maintained at 37°C, 5% CO 2 , in a humidified environment and utilized in BSL2 certified tissue culture hoods using sterile technique and appropriate personal protective equipment including goggles, gloves and lab coats.
- An 80 ⁇ L volume of cells is plated overnight in growth medium at a cell density of about 50,000 cells/mL.
- ArgRS polypeptides at a final concentration of 250 nM per well are added in sterile PBS to each well following overnight adherence.
- Control wells contain untreated cells and are incubated with an equivalent volume of PBS. Cells are exposed to proteins or PBS in basal media (Cell Applications Cat #310-470) for 24 hours. Supernatant is removed and IL-8, IL-6 and TNFa ELISA assays are run according to manufacturer's instructions (RND Systems, Cat # DY206 and DY-208, DY-210 Duo-set kits). Proliferation is assessed with Resazurin as described previously by adding fresh media containing Resazurin to plates following supernatant removal and incubating for three hours at 37 °C. Plates are read on a fluorescent plate reader and viability / proliferation is expressed as a function of resorufin associated fluorescence of ArgRS polypeptide treated wells divided by resorufin associated fluorescence of PBS only treated wells.
- HA Human astrocytes
- HA Human astrocytes
- HA are derived from human cerebral cortex. They are cryopreserved at second passage and can be cultured and propagated 10 population doublings. HA are the most abundant cells in the central nervous system and they perform many functions such as provision of mechanical support and nutrients to neurons, and removal of wastes from neurons. In addition to playing a critical support role for optimal neuronal functioning, they also provide biochemical support of endothelial cells which form the blood-brain barrier. Recent studies have shown that astrocytes are capable of regulating neurogenesis by instructing the stem cells to adopt a neuronal fate and controlling the function of single synapses, participate actively in the transfer and storage of information in the brain.
- HA can serve as useful in vitro model for exploring the diversity of astrocytes functions.
- Astrocytes have been shown to proliferate in response to IL6 and TNFalpha.
- these cells are capable of making their own IL6 and TNFalpha.
- ArgRS polypeptides which modulate the proliferation and cytokine production in HA have therapeutic utility in a variety of neurological diseases including neuro-inflammation, neurodegeneration, tumorigenesis of the brain, and brain ischemia and repair.
- HA Human Astrocytes (HA) from Cell Applications (Cat # 882K-05f) are maintained in Cell Applications HA Cell Growth Medium (Cat # 821-500) according to manufacturer's instructions.
- Cells are maintained at 37°C, 5% CO 2 , in a humidified environment and utilized in BSL2 certified tissue culture hoods using sterile technique and appropriate personal protective equipment including goggles, gloves and lab coats.
- An 80 ⁇ L volume of cells is plated on collagen coated plates overnight in complete medium (above) at a cell density of 50,000 cells/mL. Cells are washed once with PBS and 80 ⁇ L of serum free growth media is added to each well.
- ArgRS polypeptides at a final concentration of 250 nM per well are added in a consistent volume in sterile PBS to each well.
- Cells are exposed to ArgRS polypeptides for 48 hours and spent media is removed for cytokine assessment (as described previously).
- Cells are exposed to proteins or PBS in basal media (Cell Applications Cat #310-470) for 48 hours.
- Supernatant is removed and IL-8 and IL-6 ELISA assays are run according to manufacturer's instructions (RND Systems, Cat # DY206 and DY-208, DY-210 Duo-set kits).
- Proliferation is assessed with Resazurin as described previously by adding fresh media containing Resazurin to plates following supernatant removal and incubating for three hours at 37 °C. Plates are read on a fluorescent plate reader and viability / proliferation is expressed as a function of resorufin associated fluorescence of ArgRS polypeptide treated wells divided by resorufin associated fluorescence of PBS only treated wells.
- HLMVEC Human lung microvascular endothelial cell proliferation and inflammatory cytokine production
- the pulmonary vasculature is of great physiological/pathological significance. It is now recognized to be a tissue composed of metabolically active, functionally responsive cells, that interact with circulating substrates and formed elements in ways that regulate the composition of systemic arterial blood, affect target organ functions, and contribute to thrombosis, hemostasis and immune reactions, as well as tumor metastasis.
- Human lung microvascular endothelial cells (HLMVEC) exhibit elevated expression of chemoattractant cytokines and cell adhesion molecules that provide critical cues for directed migration of leucocytes into the lung during acute lung injury. This primary cell type can be useful tool for studying various aspects of pathology and biology of the pulmonary microvasculature in vitro.
- Alteration in the structure and function of the microvasculature in response to inflammatory stimuli is believed to be a key factor in organ damage and under appropriate conditions, may provide a stimulus for repair.
- a significant cause of these vascular alterations is the induction of an inflammatory reaction involving leukocyte infiltration.
- a variety of studies focused on granulocyte adhesion to the endothelium have revealed that leukocyte recruitment and emigration involves a well- orchestrated adhesion cascade. The adhesion cascade begins when the granulocyte attaches to the endothelium and begins to roll in the direction of fluid flow at a low velocity.
- ArgRS polypeptides which modulate proliferation and / or cytokine production of human lung microvascular endothelial cells have therapeutic utility in a variety of vascular and pulmonary diseases including inflammatory and obstructive lung diseases including for example, pulmonary hypertension, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and asthma.
- HLMVEC Cell Applications, Catalog # 540-05
- Cell Applications are maintained in Cell Applications Microvascular Endothelial Cell Growth Medium (Cat # 111-500)
- an Attachment Factor Solution containing collagen (Cell Applications, Catalog # 123-100)
- Cells are maintained at 37°C, 5% CO 2 , in a humidified environment and utilized in BSL2 certified tissue culture hoods using sterile technique and appropriate personal protective equipment including goggles, gloves and lab coats.
- a 80 ⁇ L volume of cells is plated on collagen coated plates overnight in complete medium (above) at a cell density of 50,000 cells/mL.
- ArgRS polypeptides at a final concentration of 250 nM per well (or as otherwise described in the examples below) are added in a consistent volume in sterile PBS to each well. Cells are exposed to ArgRS polypeptides for 48 hours and spent media is removed for ELISA for cell adhesion molecules and cytokine assessment (as described previously). Cell adhesion molecules including soluble VCAM and/or ICAM are measured using a standard ELISA kit from RND Systems (Cat # DY643 and DY720 respectively).
- Proliferation is assessed with Resazurin as described previously by adding fresh media containing Resazurin to plates following supernatant removal and incubating for three hours at 37 °C. Plates are read on a fluorescent plate reader and viability / proliferation is expressed as a function of resorufin associated fluorescence of ArgRS polypeptide treated wells divided by resorufin associated fluorescence of PBS only treated wells.
- CAMs Cell Adhesion Molecules
- ECM extracellular matrix
- CAMs are proteins located on the cell surface which are involved with the binding with other cells or with the extracellular matrix (ECM) in the process called cell adhesion. These proteins are typically transmembrane receptors and are composed of three domains: an intracellular domain that interacts with the cytoskeleton, a transmembrane domain, and an extracellular domain that interacts either with other CAMs of the same kind (homophilic binding) or with other CAMs or the extracellular matrix (heterophilic binding). Most of the CAMs belong to four protein families: Ig (immunoglobulin) superfamily (IgSF CAMs), the integrins, the cadherins, and the selectins.
- Ig immunoglobulin
- the immunoglobulin superfamily (IgSF) cell adhesion molecules are calcium-independent transmembrane glycoproteins, including: neural cell adhesion molecules (NCAMs), intercellular cell adhesion molecules (ICAMs), vascular cell adhesion molecule (VCAM), platelet-endothelial cell adhesion molecule (PECAM-1), endothelial cell-selective adhesion molecule (ESAM), junctional adhesion molecule (JAMs), nectins, and other cell adhesion molecules.
- NCAMs neural cell adhesion molecules
- ICMs intercellular cell adhesion molecules
- VCAM vascular cell adhesion molecule
- PECAM-1 platelet-endothelial cell adhesion molecule
- ESAM endothelial cell-selective adhesion molecule
- JAMs junctional adhesion molecule
- nectins and other cell adhesion molecules.
- ICAM-1 Intracellular fibroblasts
- ICAM-1 Intracellular cytoplasmic factor-1
- ICAM-1 Intracellular cytoplasmic factor-1
- ICAM-1 is of particular importance since it mediates firm endothelial adhesion and facilitates leukocyte transmigration.
- Studies have shown that there is an upregulation of ICAM-1 on both sinusoidal cells and hepatocytes in inflammatory liver conditions such as hepatitis B viral infection, autoimmune liver disorders, alcoholic hepatitis, and liver allograft rejection.
- ArgRS polypeptides which modulate cell adhesion molecule production and cell adhesion to endothelial cells have therapeutic utility in a variety of inflammatory diseases including for example, cardiovascular diseases, atherosclerosis, autoimmunity and pulmonary hypertension.
- Human umbilical vein cells (ATCC, Cat # CRL-2873) (HUVEC) are seeded at a concentration of about 1.2 x 10 5 cells / well in 12 well plates coated with human fibronectin attachment solution in the suggested ATCC media and supplements and grown according to manufacturer's instructions. Cells are stimulated with ArgRS polypeptides at the indicated concentrations, or PBS alone, and incubated overnight in growth media.
- Human acute monocytic leukemia (THP-1 (TIB-202) cells are resuspended into 0.1% BSA/ RPMI serum free medium with calcein AM (6 ⁇ L/mL; Invitrogen Cat # C1430) and incubated for 30 minutes. Labeled cells are collected and resuspended in RPMI medium containing 10 % FBS, and the density adjusted to 2 x 10 6 cells/mL.
- THP-1 cells 100 ⁇ L (2 x 10 5 ) labeled THP-1 cells are placed into each well of the HUVEC monolayer in 1 mL of growth media and incubated for 15 minutes. The wells are washed twice with PBS to remove unbound cells, and then the cells are read by fluorescent plate reader with an Excitation wavelength of 488 nm and an Emission wavelength of 530 nm.
- Adipocyte differentiation and proliferation in primary human pre - adipocyte cells Adipocyte differentiation and proliferation in primary human pre - adipocyte cells.
- adipose tissue is an endocrine organ that secretes a wide variety of factors, and dysregulated secretion affects adipogenesis as well as whole-body glucose/insulin homeostasis. Excess adipose tissue leading to obesity has become a severe public health threat. Adipose tissue development can be affected by genetic background, hormonal balance, diet, and physical activity. Adipose tissue mass can increase when fat cells are increased in size due to higher triacylglycerol accumulation.
- adipocytes specifically are thought to arise from mesenchymal cells that undergo the commitment and differentiation process, adipogenesis.
- Pre-adipocyte cell lines can undergo adipocyte differentiation upon treatment with adipogenic agents comprised of synthetic glucocorticoid, dexamethasone (DEX), isobutylmethylxanthine (IBMX), and insulin, have been valuable in these studies.
- adipogenic agents comprised of synthetic glucocorticoid, dexamethasone (DEX), isobutylmethylxanthine (IBMX), and insulin
- Peroxisome proliferator-activated receptor ⁇ (PPAR ⁇ ) and CCAAT enhancer-binding protein (C/EBP) family of transcription factors have been firmly established to play critical roles in adipocyte differentiation.
- C/EBP ⁇ and C/EBP ⁇ are induced by DEX and IBMX, respectively, which together then induce PPAR ⁇ and C/EBP ⁇ to activate various adipocyte markers that are required for adipocyte function.
- Other transcription factors have also been reported to either positive or negatively regulate adipogenesis and various growth factors and hormones can affect adipocyte differentiation by regulating expression of adipogenic transcription factors.
- adipose tissue secretes a wide array of molecules that are involved in diverse physiological processes including immune response, vascular function, and energy homeostasis.
- Cytokines such as TNF- ⁇ and IL-6 are secreted from adipocytes. Some of these factors may also affect growth and development of adipose tissue by autocrine/paracrine action.
- ArgRS polypeptides which have the ability to modulate the differentiation and / or proliferation of normal human pre-adipocytes have therapeutic utility in a broad range of diseases including for example, the treatment and prevention of metabolic disease, cardiovascular diseases, obesity and lipodystrophies, as well as the long term complications of diabetes.
- HPAd human pre-adipocytes
- Cell Application Cat # 803sD HPAd (human pre-adipocytes)
- HPAd human pre-adipocytes
- cells are thawed quickly, and transferred immediately into 15mL of Adipocyte Growth Medium (Cell Application Cat # 811M-250) and plated into a standard sterile tissue culture treated flask. Media is replaced with fresh Adipocyte Growth Medium every other day until cell is >60% confluent.
- Cells are grown at 37°C, 5% CO 2 , in a humidified environment and utilized in BSL2 certified tissue culture hoods using sterile technique and appropriate personal protective equipment including goggles, gloves and lab coats.
- Cells are plated in clear bottom black walled 96 well tissue culture treated assay plates for differentiation at a concentration of about 50,000 cells/mL.
- ArgRS polypeptides at a final concentration of 250nM per well (or as otherwise indicated in the Examples below) are added to each assay well. All cells are maintained in growth media for 2 days with the exception of the positive controls which are stimulated with adipogenic differentiation media (Cell Applications Cat #811D-250). Cells are exposed to ArgRS polypeptides for 48 hours.
- Cell adhesion molecules including soluble VCAM and/or ICAM are measured using a standard ELISA kit from RND Systems (Cat # DY643 and DY720 respectively).
- Proliferation is assessed with Resazurin as described previously by adding fresh media containing Resazurin to plates following supernatant removal and incubating for three hours at 37 °C. Plates are read on a fluorescent plate reader and viability / proliferation is expressed as a function of resorufin associated fluorescence of ArgRS polypeptide treated wells divided by resorufin associated fluorescence of PBS only treated wells. Fresh media is added and differentiation is maintained for 16 days post initial media exchange, with fresh media exchanged every other day to maintain cell health. On day 15, cells are placed in serum free media.
- differentiation to mature adipocytes is assessed with Nile Red (Invitrogen, concentration of 3 ⁇ M final) staining and quantified with a fluorescent plate reader with the appropriate wavelengths.
- Nile Red Invitrogen, concentration of 3 ⁇ M final
- cells are fixed with 10% paraformaldehyde, washed in PBS and permeabilized in PBS containing 0.5% BSA and 0.1% Triton X-100.
- Cell proliferation is assessed with an intensity measurement on a fluorescent reader with Hoechst dye 33432 at a concentration of 1ug/mL final, as described previously.
- Adipogenesis is expressed as intensity of Nile Red signal. Hoechst dye signal is used to assess cellular number.
- skeletal muscle is a multistep process that involves the determination of pluripotential mesodermal cells to give rise to myoblasts, withdrawal of the myoblasts from the cell cycle and differentiation into muscle cells, and finally growth and maturation of skeletal muscle fibers.
- Skeletal muscle differentiation involves myoblast alignment, elongation, and fusion into multinucleate myotubes, together with the induction of regulatory and structural muscle-specific genes.
- myogenic commitment and muscle-specific gene expression involve the skeletal muscle-specific helix-loop-helix (bHLH) MyoD family of proteins, which includes MyoD, myogenin, myf-5, and MRF4, and the myocyte enhancer-binding factor 2 (MEF2).
- MyoD family proteins The DNA binding activity of MyoD family proteins is attenuated by Id, which forms complexes with E2a gene products in proliferating cells and is down-regulated when they are induced to differentiate.
- the decision to differentiate into myotubes is influenced negatively by several factors.
- Treatment of myoblasts with fetal bovine serum, basic fibroblast growth factor 2, or transforming growth factor ⁇ 1 is known to inhibit differentiation of myoblasts.
- Myogenesis is also regulated negatively by oncogenes such as c-myc, c-jun, c-fos, H-ras, and E1a. There is very little information regarding the signaling that is triggered in the myoblast upon serum withdrawal which leads to the induction of the MyoD family gene expression and to muscle differentiation.
- IGF insulin-like growth factor
- ArgRS polypeptides with the ability to modulate muscle development have therapeutic utility in a broad range of diseases including for example, the treatment of metabolic disease, cachexia, various muscle wasting conditions, as well as musculoskeletal disease where muscle atrophy plays a key role in the pathogenesis and symptomology.
- Human Skeletal Muscle Cells (HSkMC) can undergo differentiation to exhibit actin and myosin myofilaments.
- HSkMC have been used in the study of genetic muscular diseases such as Malignant Hyperthermia.
- HSkMC also have the potential to act as a cardiac graft, mending damage to the heart, and thus ArgRS polypeptides with the ability to modulate muscle development also have utility as in vitro and in vivo regulators of myogenesis.
- HSkMC Human Adult Skeletal Muscle Cells
- Cell Applications Cat # 151-500
- HSkMC Growth Medium Cell Applications, Cat # 151-500
- These cells can be cultured and propagated for at least 15 population doublings.
- For differentiation cells are maintained in growth media for one passage and then plated at 50,000 cells per mL media in to 96 well clear bottom black walled TC treated plates treated with collagen at 100 ⁇ L per well. Cells are allowed to adhere overnight.
- ArgRS polypeptides in PBS, or PBS alone, is added to each well at a final concentration of 250nM protein (or as otherwise indicated in the examples below).
- Control wells received the same volume of Differentiation Media (Cell Applications Cat # 151D-250) at this time. Cells are incubated with protein or differentiation media for 48 hours. At 48 hours, cell culture supernatant is collected from all wells and differentiation media is added at a volume of 150 ⁇ L to the entire plate with the exception of control wells which are maintained in growth media only. Supernatant is utilized to assess cytokine production including IL6 and IL8 as described previously.
- Proliferation is assessed with Resazurin as described previously by adding fresh media containing Resazurin to plates following supernatant removal and incubating for three hours at 37 °C.
- Cells are monitored under the microscope and media is exchanged for fresh Differentiation media every 2 days. On Day 10, media is removed and cells are fixed with 10% paraformaldehyde for 30 minutes.
- Cells are permeabilized with 0.1% Triton X-100 in PBS for 15 minutes and cells are stained with TR-Labeled phalloidin and Hoechst 33432 (as described previously) to define actin and nuclei respectively. Nuclear intensity is used to determine cell proliferation in each well and phalloidin intensity is used to determine total actin content.
- Cells are also stained with alpha actin skeletal muscle antibody (GenTex Cat # GTX101362). Digital photos using a fluorescent microscope as well as visual inspections and scoring are made of all wells.
- MSCs Mesenchymal stem cells
- MSCs are multipotent stem cells that can differentiate into a variety of cell types, including osteoblasts, chondrocytes, myocytes, adipocytes, beta-pancreatic islets cells, and potentially, neuronal cells.
- Many different events contribute to the commitment of the MSC to other lineages including the coordination of a complex network of transcription factors, cofactors and signaling intermediates from numerous pathways.
- MSCs are of intense therapeutic interest because they represent a population of cells with the potential treat a wide range of acute and degenerative diseases.
- ArgRS polypeptides with the ability to modulate the differentiation of MSCs into different developmental pathways have significant therapeutic utility to enable the in vitro or in vivo modulation of hematopoiesis, neurogenesis, myogenesis, osteogenesis, and adipogenesis, as well as in a broad range of disorders and diseases, including for example inflammatory responses, autoimmunity, cancer, neuronal degeneration, muscular dystrophy, osteoporosis, and lipodystrophy.
- Human MSCs are immuno-privileged, and represent an advantageous cell type for allogenic transplantation, reducing the risks of rejection and complications of transplantation.
- MSCs for gene therapy, including transplantation of MSCs transfected with vascular endothelial growth factor for the improvement of heart function after MI in rats, MSCs as vehicles for interferon- ⁇ delivery into tumors in mice and gene therapy with MSCs expressing BMPs to promote bone formation.
- ArgRS polypeptides were tested as potential inducers of MSC proliferation and differentiation.
- hMSC human marrow stromal cells
- Cell Application Cat # 492-05f human marrow stromal cells
- hMSC human marrow stromal cells
- Cell Application Cat # 419-500 Marrow Stromal cell Growth Medium
- Media is replaced with fresh Marrow Stromal cell Growth Medium every other day until cells are >60% confluent.
- Cells are grown at 37°C, 5% CO 2 , in a humidified environment and utilized in BSL2 certified tissue culture hoods using sterile technique and appropriate personal protective equipment including goggles, gloves and lab coats.
- Cells are plated in clear bottom black walled 96 well tissue culture treated assay plates for differentiation at a concentration of 50,000 cells/mL.
- tRNA synthetase derived proteins at a final concentration of 250 nM per well (or as otherwise specified in the Examples below) are added to each assay well. All cells are maintained in growth media for 2 days with the exception of the positive controls, which was stimulated with osteogenic or chonodrogenic differentiation media (StemPro, Invitrogen, Cat # A10072-01 and A10071-01 respectively).
- Cells are exposed to ArgRS polypeptides for 48 hours. Soluble VCAM is measured using a standard ELISA kit from RND Systems (Cat # DY643).
- Proliferation is assessed with Resazurin as described previously by adding fresh media containing Resazurin to plates following supernatant removal and incubating for three hours at 37 °C. Plates are read on a fluorescent plate reader and viability / proliferation is expressed as a function of resorufin associated fluorescence of ArgRS polypeptide treated wells divided by resorufin associated fluorescence of PBS only treated wells. Following an assessment of cell viability, resazurin is removed with two media exchanges and 0.5X differentiation media is added to all wells. Differentiation is monitored by visual inspections of all wells for 10 days post media exchange, with fresh media exchanged every other day to maintain cell health.
- hPASMC Human pulmonary artery smooth muscle cell proliferation and differentiation .
- Pulmonary artery smooth muscle cells in normal human adult lung blood vessels are mostly quiescent, non-migratory and are largely committed to executing their contractile function in the lung.
- PASMCs are not terminally differentiated and possess the ability to modulate their phenotype and exit their quiescent state in response to changing local environmental cues. This differentiation state may occur in development, tissue injury, and vessel remodeling in response to changes in tissue demand.
- Pulmonary hypertension PH is associated with a variety of underlying conditions including an increase in peripheral pulmonary vascular resistance as a result of increased vascular tone and PASMC contractility and vascular remodeling.
- Vascular remodeling involves PASMC growth, synthesis of matrix material, and alterations in cell-cell and cell-matrix interactions in the walls of small pulmonary arteries (PAs), which lead to increased thickness of the smooth muscle component of the vessel wall and abnormal muscularization of the normally nonmuscularized, distal PAs. This process contributes to reduced lumen diameter and increased peripheral resistance.
- PAs small pulmonary arteries
- a crucial step in studying cellular differentiation is identifying a set of cell-specific or cell-selective genes that contribute to the differentiated function(s) of the cell.
- SMC smooth muscle cell
- ArgRS polypeptides which have the ability to modulate the differentiation and / or proliferation of normal human PASMCs derived from adult humans have therapeutic utility in a variety of vascular and pulmonary diseases including inflammatory and obstructive lung diseases including for example, pulmonary hypertension, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and asthma.
- HPASMC Cell Applications Cat # 352-05a
- HPASMC growth media Cell Applications Cat # 352-05a
- Cells are maintained at 37°C, 5% CO 2 , in a humidified environment and utilized in BSL2 certified tissue culture hoods using sterile technique and appropriate personal protective equipment including goggles, gloves and lab coats.
- An 80 ⁇ L volume of cells is plated on collagen coated overnight in growth medium at a cell density of 50,000 cells/mL.
- ArgRS polypeptides were added in sterile PBS to each well at a final concentration of 250 nM (or as otherwise specified in the Examples below). Control wells held only an equivalent volume of PBS.
- Positive control samples were incubated with vendor supplied HPASMC differentiation media (Cell Applications Cat # 311D-250). Cells are exposed to ArgRS polypeptides or PBS in basal media (Cell Applications Cat # 310-470) for 48 hours followed by a media exchange to differentiation media for the entire plate. Supernatant is collected and utilized to assess cytokine production including IL6 and IL8 as described previously. Proliferation is assessed with Resazurin as described previously by adding fresh media containing Resazurin to plates following supernatant removal and incubating for three hours at 37 °C. Cells are monitored for 10 days with a media exchange every other day.
- Triton X-100 Differentiation is assessed after fixation as described above, and permeabilzation with 0.1% Triton X-100, by quantifying staining to smooth muscle actin-alpha staining using an anti-SMA-alpha antibody (GeneTex Cat #GTX101362) and an Alexa 405 conjugated secondary antibody. Proliferation is assessed with Hoechst staining after cell fixation in 10% formaldehyde for 30 minutes. Hoechst dye is read using a bottom reading fluorescent plate reader with an excitation wavelength (Ex) of 405 nm, and an emission wavelength (Em) of 450 nm. Total actin staining is assessed via the use of an Alexa-488 labeled phalloidin stain (Invitrogen Cat# A12379).
- ArgRS polypeptide binding demonstrates that the cell type in question expresses specific receptors for the ArgRS polypeptide in question.
- cell binding implies a potential role for the ArgRS polypeptide in regulating the activity or behavior of the cell, or similar types of cell, in vivo.
- regulatory roles include for example, the binding and modulation of B-cells and T-cells (immunomodulation / chemotaxis / autoimmunity / inflammation); HepG2 cells (control of metabolism, cholesterol uptake or metabolism); THP-1, Jurkat, Raji cells (immunomodulation / chemotaxis / autoimmunity / inflammation), platelets (thrombopoiesis), 3T3L1 adipocytes (lipogenesis / metabolism), and C2C12 mouse myoblasts (myogenesis, osteogenesis).
- Blood is collected in EDTA tubes from healthy donors. 2mL whole blood is placed into 5mL Falcon FACS tube. 2mL of staining buffer (PBS + 2% FBS) is added, vortexed 3-5 seconds, centrifuged for 5 minutes at 300 x g. The supernatant aspirated, the wash repeated, and the pellet resuspended in 2 mL of staining buffer.
- staining buffer PBS + 2% FBS
- Platelet binding assays 50 ⁇ L of washed blood is transferred to clean 5mL FACS sample tubes, His6- or V5-His6-tagged ArgRS polypeptides are added to tubes at the concentrations indicated in the specific experiments outlined below and tubes are placed on ice for 45 minutes. 20 ⁇ L CD61 pan platelet antibody (BD Pharmigen, Cat # 555754) and 0.5 ⁇ L anti- V5-FITC labeled antibody (Invitrogen, R96325) or FITC labeled anti-His6 antibody (AbCam Cat #ab1206) are added to each tube. Tubes are placed on ice and protected from light for 30 minutes.
- Binding to cells in culture Approximately 1 x 10 6 cells in 100 ⁇ L complete RPMI medium are placed into 5mL FACS tubes. His6- or V5-His6-tagged ArgRS polypeptides are added to tubes at the concentrations indicated in the specific experiments outlined below and tubes are placed on ice for 45 minutes. Cell samples are washed twice with 1mL staining buffer (PBS + 2% FBS), and then 0.5 ⁇ L of anti-V5-FITC antibody (Invitrogen R96325) or FITC labeled anti-His6 antibody (AbCam Cat #ab1206) in staining buffer with 200 ⁇ g/mL human IgG, is added and the samples incubated on ice, protected from light, for 30 minutes.
- staining buffer PBS + 2% FBS
- ANIMAL STUDIES MODULATION OF HAEMATOPOIESIS AND CIRCULATING CYTOKINES
- Hematopoiesis is the formation of blood cellular components. All cellular blood components are derived from haematopoietic stem cells (HSCs) which reside in the medulla of the bone (bone marrow) and have the unique ability to give rise to all of the different mature blood cell types. HSCs are self-renewing: when they proliferate, at least some of their daughter cells remain as HSCs, so the pool of stem cells does not become depleted.
- HSCs haematopoietic stem cells
- HSCs myeloid and lymphoid progenitor cells
- ArgRS polypeptide is capable of modulating hematopoiesis, and regulating the development of haematopoietic stem cells.
- All blood cells can be divided into three lineages; Erythroid cells, lymphocytes and myelocytes.
- Erythroid cells are the oxygen carrying red blood cells. Both reticulocytes and erythrocytes are functional and are released into the blood. Accordingly a reticulocyte count estimates the rate of erythropoiesis, and a change in red blood cell count suggests that an ArgRS polypeptide modulates erythropoiesis.
- Lymphocytes are the cornerstone of the adaptive immune system. They are derived from common lymphoid progenitors. The lymphoid lineage is primarily composed of T-cells and B-cells (types of white blood cells). Accordingly a change in white blood cell count or composition in response to exposure to an ArgRS polypeptide suggests that that the ArgRS polypeptide modulates lymphopoiesis.
- Myelocytes which include granulocytes, megakaryocytes and macrophages, and are derived from common myeloid progenitors, are involved in a variety of roles, including innate immunity, adaptive immunity, and blood clotting. Accordingly a change in myeloid cell count or composition in response to exposure to an ArgRS polypeptide suggests that that the ArgRS polypeptide modulates myelopoiesis. The same rationale can be used to establish whether the ArgRS polypeptides modulate granulopoiesis, by measuring changes in granulocyte number in response to exposure to the ArgRS polypeptides. A role for the ArgRS polypeptide in modulating megakaryocytopoiesis may be inferred by a change in megakaryocyte or platelet composition or number in the blood.
- Cytokine release in either wild type mice, or in various animal model systems of inflammation provides an initial assessment of the potential ability of the ArgRS polypeptides to modulate inflammatory responses.
- the role of ArgRS polypeptides in modulating acute chronic inflammatory processes for example, can be readily assessed using a mouse model of diet induced obesity (DIO).
- DIO diet induced obesity
- the DIO model centers upon placing rodents on a high fat diet for several months leading to increased obesity, insulin resistance and immune system dysfunction.
- a particular consequence of this immune system dysregulation results in increased production of proinflammatory cytokines in DIO animals leading to a condition of chronic systemic inflammation.
- ArgRS polypeptides to modulate the immune system and restore homeostatic balance towards a resolution of this chronic inflammatory state would be particularly beneficial in numerous diseases and disorders including but not limited to the treatment and prevention of the symptoms and side effects of metabolic disease, diabetes, cardiovascular diseases, atherosclerosis, obesity, as well as various autoimmune diseases and disorders, including for example, multiple sclerosis, vascular and allergic disorders.
- mice Male wild type control (C57BL/6) or diet induced obesity mice (C57BL/6NHsd) are purchased from Harlan (Indianapolis, IN) and housed individually. DIO mice are fed a high fat diet (Cat. #TD.06414-60% kcal from fat) and control mice are fed a normal diet (Cat. #2018S-18% kcal from fat). DIO mice are placed on the high fat diet starting at 6 weeks of age for a total of 10 weeks. Both DIO and control mice are allowed to feed and drink ad libitum. At 16 weeks of age, mice are sorted and randomized into groups of 5 animals based on weight. On day 2, mice are weighed and tail vein bled (100 ⁇ L) for pre-treatment complete blood count (CBC) analysis.
- CBC complete blood count
- mice are weighed and intravenously injected via the tail vein with vehicle (PBS) or individual ArgRS polypeptides at 10mg/kg.
- PBS vehicle
- mice are facial vein bled (150-200 ⁇ L) for subsequent cytokine analysis.
- mice are intravenously dosed as on day 1.
- mice are weighed, terminated and blood are collected by heart puncture for Complete Blood Count (CBC analysis) (plasma-EDTA) and cytokine examination (serum).
- CBC analysis Complete Blood Count
- CBC and cytokine analysis Complete blood counts are analyzed from blood draws preceding injections (day -2) and 24 hours after the final injection (day 5). CBC values are assessed for total white blood cell counts and overall red blood cell morphology. White blood cells are further characterized by total and fractional percentage of neutrophils, lymphocytes, monocytes, eosinophils, & basophils. Red blood cell breakdown included measurements of hemoglobin (dL), hematocrit (%), mean corpuscular volume (fL), mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration (%), and total platelet count (10 3 / ⁇ L). CBC analysis is performed by Antech Diagnostics (Fishers, IN).
- Circulating cytokine levels are examined at 4 hours post-injection (day 1) and 24 hours after the final injection (day 5). Serum is isolated, snap frozen and sent to Rules Based Medicine (Austin, TX) for multi-analyte profiling. Serum samples are analyzed using the RodentMap panel encompassing 59 unique biomarkers including Apo A-1, CD40, CD40-L, CRP, ET-1, eotaxin, EGF, Factor VII, fibrinogen, FGF-9, FGF-basic, GST- ⁇ , GCP-2, GM-CSF, KC/GRO ⁇ , haptoglobin, IgA, IFN ⁇ , IP-10, IL-1 ⁇ , IL-1 ⁇ , IL-10, IL-11, IL-12p70, Il-17A, IL-18, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, LIF, lymphotactin, M-CSF-1, MIP-1 ⁇ , MIP-1 ⁇ , MIP-1 ⁇ ,
- ArgRS fragments from cell lines, conditioned media and tissues, samples are prepared in the following ways:
- Mouse macrophage (RAW 264.7), cytosol and conditioned media Cells are treated with serum free DMEM media at a density of 15 x 10 6 cells / flasks. After 48 hours conditioned media and cell pellets are collected and processed. 200 ⁇ g of protein from secreted and cytosolic proteomic fractions are separated by SDS-PAGE and gel slices are prepared for analysis by mass spectrometry.
- Mouse pancreas tissue The pancreas from three mice are chopped, dounce homogenized, and sonicated in PBS with protease inhibitors. Cytosolic proteome is isolated by centrifugation and 200 ⁇ g of protein is separated by SDS-PAGE and gel slices are prepared for analysis by mass spectrometry.
- Mouse liver tissue Three mouse livers are chopped, dounced homogenized, and sonicated in PBS with protease inhibitors. Cytosolic proteome is isolated by centrifugation and 200 ⁇ g of protein is separated by SDS-PAGE and gel slices are prepared for analysis by mass spectrometry.
- In-gel digests are analyzed by LTQ XL ion trap mass spectrometer (ThermoFisher) equipped with ultimate 3000 ⁇ LC system (Dionex).
- the samples are first loaded on PepTrap (michrom) for 10 min with 5% Acetonitrile in 0.1% formic acid using Dionex autosampler. Then the samples are analyzed with a 100 ⁇ m (inner diameter) fused silica capillary column containing 10 cm of C18 resin (michrom). Peptides are eluted from the column into mass spectrometer with a flow rate of 0.45 ⁇ l/min using a linear gradient of 5-33.5% acetonitrile in 0.1% formic acid within 110 min.
- LTQ is operated in data-dependent scanning mode such that one full MS scan is followed by seven MS/MS scans of the seven most abundant ions.
- Dynamic exclusion is enabled with repeat count equals to 1, repeat duration equals to 20 seconds, exclusion list size is 300 and exclusion duration is 60 seconds.
- the raw data is searched with BioWorks3.3.1(SEQUEST) using a concatenated target/decoy variant of the mouse IPI database.
- SEQUEST data are filtered and sorted with DTASelect. Tables 1, 4 and 7 show sequences identified in this way.
- Splice variants of the arginyl aminoacyl tRNA synthetase are identified using high throughput sequencing of cDNA libraries enriched for arginyl aminoacyl tRNA synthetase transcripts.
- the cDNA templates are prepared from total RNA extracts of tissues such as human adult and fetal brains and enriched for aminoacyl tRNA synthetase transcripts by using primer sequences specific for all annotated exons of all annotated human aminoacyl tRNA synthetases and their associated proteins.
- RNA samples Human Total RNAs are obtained from Clontech. For cell line and mouse tissue samples, total RNAs are extracted using RNA Extract II Kit (MN). Genomic DNA is digested in the total RNA samples by DNAase I. To obtain mature messenger RNAs (mRNAs), the RNA samples are enriched twice by binding polyA+ RNA and digestion of RNA without 5'-cap by 5'-phosphate dependent exonuclease. Complementary DNA (cDNA) is synthesized from mature RNAs using primers that anneal to exon sequences of aminoacyl tRNA synthetase genes.
- cDNA Complementary DNA
- a transcriptome enriched for aminoacyl tRNA synthetase genes is amplified by multiplex PCR using the aminoacyl tRNA synthetase-exon specific cDNA and different combinations of aminoacyl tRNA synthetase-exon primers.
- the double-stranded aminoacyl tRNA synthetase-enriched transcriptome PCR products are enzymatically repaired at both ends before adding A-overhangs to the 3' ends of the repaired fragments.
- Sequencing adaptors and index sequences are then added to the aminoacyl tRNA synthetase-enriched transcriptome PCRs products to generate cDNA libraries for deep sequencing with Illumina's Multiplex Sequencing Kit.
- the aminoacyl tRNA synthetase-enriched transcriptome PCR products with 3'-A overhangs are ligated to the InPE adaptor oligonucleotides provided in the kits.
- Index sequences are added to the PCR products with InPE adaptors.
- the PCR products with index sequences are further amplified by PCR.
- Aminoacyl tRNA synthetase-enriched cDNA libraries with different indexes are pooled and sequenced using an Illumina DNA sequencing machine to get 50 base pair end reads. Sequencing reads are mapped to human or mouse genome for identification of alternative splicing events.
- "Splicemap" software (available for public download at http://www-stat.stanford.edu/-kinfai/SpliceMap/) is used to identify splice junctions.
- Deep sequencing of these cDNAs are performed to generate about 1 million sequencing reads of about 50 nucleotides in length.
- the sequences specific for exons of the aminoacyl tRNA synthetases are queried against annotated exon junctions and new exon junctions are identified as alternative splicing events.
- Tables 2 and 5 labeled "5' exon” and "3'exon” indicate, when present, which exons are fused together in the cDNA sequence.
- Tables 2 and 5 show sequences that were identified for alternative splice events, transcripts containing such splice events, and the polypeptides expressed by those transcripts.
- Alternative splice variants identified by deep sequencing are identified in Tables 2 and 5 as those ones in which there are numbers greater than zero in the columns labeled as "Sequencing reads" in the human adult or fetal brain.
- FASTA available at the website http://fasta.bioch.virginia.edu/fasta_www2/
- Resectin sequences from the human proteins are identified as sequences covering regions where there are gaps in the bacterial sequence in the alignment, or regions with low homology between the two species.
- the peptide, and corresponding DNA sequences in Tables 3 and 6 include examples identified in this way.
- the PROTOMAP technique is used as described in Example 1 to compare the differential expression of Arginyl tRNA synthetases in different tissues/cell types (refer to Tables 1, 4, and 7 for sequences and comparisons): Aminoacyl-tRNA synthetase resectin expression is compared between mouse liver tissue and mouse pancreas tissue. Aminoacyl-tRNA synthetase resectin expression is compared between cytosol of RAW264.7 and conditioned media from RAW264.7 cells harvested after 48 hours of serum starvation.
- a human antibody phage display library is screened by AbD Serotec (a division of MORPHOSYSTM, Martinsried/Planegg, Germany) using affinity enrichment techniques (panning).
- AbD Serotec a division of MORPHOSYSTM, Martinsried/Planegg, Germany
- affinity enrichment techniques panning
- Antibodies enriched after multiple rounds of screening with the arginyl aminoacyl tRNA synthetase fragments are subsequently characterized by ELISA for reactivity to the fragments, and to the parental, full length enzyme.
- Clones demonstrating preferential binding (e.g ., ⁇ 10-fold higher affinity) to the arginyl aminoacyl tRNA synthetase fragments are further characterized.
- subtraction strategies such as pre-adsorption steps with the full length enzyme and/or counter-screening, are used to eliminate cross reacting antibodies and drive the selection process towards the unique epitope(s) on the arginyl aminoacyl tRNA synthetase fragments.
- cDNA templates for PCR reactions are reverse transcribed from total RNA extracts of tissues or cells (e.g ., human brain, IMR-32 and HEK293T).
- PCR reactions are performed using arginyl aminoacyl tRNA synthetase specific primers, pairing a forward primer (FP1) designed to anneal to the 5' untranslated region or exons in the 5' half of the gene with a reverse primer (RP1) designed to anneal to exons in the 3' half of the gene or the 3'UTR.
- FP1 forward primer
- RP1 reverse primer
- Amplified DNA products are analyzed by agarose gel electrophoresis to identify PCR products that are a different size then the fragment amplified form the canonical transcripts.
- ArgRS polypeptides are selected for further biochemical, biophysical and functional characterization based on one or more of the following criteria, i) the identification of ArgRS polypeptide proteolytic fragments, ii) the identification of ArgRS polypeptide splice variants, iii) the identification of ArgRS polypeptides by bioinformatic analysis, iv) evidence of differential expression of specific ArgRS polypeptides, v) the domain structure of the ArgRS protein, vi) the size of the ArgRS polypeptide, and vii) the minimization of similar duplicative sequences.
- Polynucleotides encoding the selected ArgRS polypeptides listed in Table E2, along with the appropriate N or C-terminal epitope tag, are synthesized and cloned as described in the General Materials and Methods section using the gene synthesis methodology listed in Table E2.
- ArgRS polypeptides listed in Table E2 are expressed in E. coli. as described in the General Materials and Methods section. The relative expression of soluble and inclusion body localized ArgRS polypeptides is summarized in Table E3 below.
- Table E3 Summary of ArgRS Polypeptide Bacterial Expression Characteristics ArgRS Polypeptide Location of Epitope Tag Amount of Protein Recovered from Soluble Fraction Amount of Protein Recovered from Inclusion Bodies ArgRS N1 N-terminal + ND ArgRS N1 C-terminal + ND ArgRS N2 N-terminal +++++ ND ArgRS N2 C-terminal +++++ ND ArgRS N3 N-terminal + ND ArgRS N3 C-terminal + ND ArgRS N4 N-terminal + ND ArgRS N4 C-terminal + + ArgRS C1 N-terminal + ND ArgRS C1 C-terminal + ND ArgRS C2 N-terminal + ++ ArgRS C2
- the protein expression data demonstrates the existence of at least one protein domain that exhibits high level expression of soluble protein when expressed in E. coli.
- the data demonstrates that the ArgRS polypeptide ArgRS1 N2 , (amino acids 1-71), defines the boundary of a novel protein domain that is highly expressed in E. coli.
- ArgRS polypeptides are prepared in larger amounts to enable further functional and biophysical characterization.
- the ArgRS polypeptides listed in Table E4 are expressed in E. coli. in large scale culture as described in the General Materials and Methods section. The yields, and specified biophysical characteristics, for each expressed soluble protein are summarized below in Table E4.
- ArgRS polypeptides were incubated with mesenchymal stem cells or human skeletal muscle cells for the times and at the concentrations shown in Table E5.
- Table E5 Transcriptional profiling of representative ArgRS Polypeptides in Mesenchymal Stem Cells (MSC) or Human Skeletal Muscle Cells (HSkMC) Test Sample Description Cell type and Exposure Time ArgRS Polypeptides Location of Epitope Tag Concentration nM MSC 24 hours MSC 72 hours HSkMC 24 hours HSkMC 72 hours ArgRS N2 N-terminal 250 4 3 3 2 ArgRS N2 C-terminal 250 1 1 5 7 ArgRS C2 N-terminal 250 2 5 5 10 ArgRS C2 C-terminal 250 0 4 7 3 ArgRS I1 N-terminal 250 2 5 7 3 ArgRS I1 C-terminal 250 5 3 10 5 Controls Average across all ArgRS polypeptides screened 3 5 6 7 Oste
- the numbers in each column represent the number of genes which were modulated, either positively or negatively by at least 4 fold compared to the control genes, as described in the general methods section.
- the data shows that specific forms of the ArgRS polypeptides tested have the surprising ability to regulate the transcription, and hence potentially modulate the developmental fate or differentiation status when added to either Mesenchymal Stem Cells (MSC) and / or Human Skeletal Muscle Cells (HSkMC). Shaded cells with bolded numbers in the table represent examples where the ArgRS polypeptide exhibits a significant impact on the regulation of gene transcription in the cell lines and times indicated in the table.
- ArgRS1 N2 , ArgRS1 C2 , and ArgRS1 I1 appear to be major regulators of Mesenchymal Stem Cell and / or human skeletal muscle cell gene expression.
- ArgRS polypeptides were incubated with the cell types, and the conditions provided in the general methods section, and Tables E5 and E6.
- ArgRS polypeptides were considered to be proliferative if greater than 3 SD away from the PBS value in the positive direction.
- a tRNA synthetase derived ArgRS polypeptide was considered to be cytotoxic if greater than 3 SD away from the PBS value in the negative direction.
- a cytotoxic compound was utilized as a negative control and the average value for this was always greater than 3 SD away from PBS average value.
- Cellular differentiation and phenotype assays Assay Description Assay Number Human hepatocyte (HepG2C3a cells) acetylated LDL uptake B1 Data analysis for ac-LDL uptake assay was performed by dividing the numerical value in the assay well by the average PBS value for the assay plate.
- ArgRS polypeptides were considered to be a modulator of ac-LDL uptake if greater than 2 SD away from the PBS value in the positive or negative direction. A visual check to confirm plate reader results was made using a fluorescent microscope.
- Human Neutrophil assays Assay Description Assay Number Neutrophil Elastase C1 Neutrophil oxidative burst (agonist) C2 Neutrophil oxidative burst (antagonist) C3 Table E6 Key to Assays and criteria for indicating a hit Data analysis for neutrophil assays was performed by dividing the numerical value in the assay well by the average PBS value for the assay plate.
- ArgRS polypeptides were considered to be a modulator of neutrophil elastase production or oxidative burst biology if greater than 2 SD away from the PBS value in the positive or negative direction.
- ArgRS polypeptides were considered to be a modulator of TLR specific biology if greater than 3 SD away from the PBS value in the positive or negative direction. Positive controls, including LPS and detection reagent were always significantly distinct and > 3 SD from PBS average value.
- Cytokine Release Assay Number Human Synoviocyte cytokine production (IL6 release) E1 Human pulmonary artery smooth muscle cell (hPASMC) cytokine production (IL6 release) E2 Human skeletal muscle cell (hSKMC) cytokine production (IL6 release) E3 Human Astrocyte cytokine production (IL6 release) E4 Whole blood IL6 release E5 Human pulmonary artery smooth muscle cell (hPASMC) cytokine production (IL8release) 72 h Incubation E6 IL8 production Assay Description Assay Number Human Synoviocyte cytokine production (IL8 release) E7 Human pulmonary artery smooth muscle cell (hPASMC) cytokine production (IL8release) E8 Human skeletal muscle cell (hSKMC) cytokine production (IL8 release) E9 Human Astrocyte cytokine production (IL8 release) E10 Human hepatocyte (HepG2C3a cells) IL8 release E11 Human acute promyelocytic leukemia
- ArgRS polypeptides were considered to be a modulator of cytokine production or cytokine related biology if the measured value was greater than 2 SD away from the PBS value in the positive or negative direction.
- a protein standard (specific to each assay kit) was run on every plate to insure good assay quality. Only assays with protein standard curves that had an R 2 value of > than 0.9 were chosen for data analysis.
- ArgRS polypeptides were considered to be a modulator of cell adhesion or a regulator of biology related to cell adhesion if the measured value was greater than 2 SD away from the PBS value in the positive or negative direction.
- a protein standard (specific to each assay kit) was run on every plate to insure good assay quality. Only assays with protein standard curves that had an R 2 value of > than 0.9 were chosen for data analysis.
- hPAD Human pre-adipocyte
- hSKMC Human skeletal muscle
- hMSC Human mesenchymal stem
- hPASMC Human pulmonary artery smooth muscle cell
- G4 Data analysis for cellular differentiation assays was performed by dividing the numerical value in the assay well by the average PBS value for the assay plate. Differentiation assays were scored based upon fluorescent intensity of particular antibodies as described in the methods section. ArgRS polypeptides were considered to be a modulator of cellular differentiation if an intensity value for a specific marker of differentiation was greater than 2 SD away from the PBS value in the positive or negative direction in a given treated well.
- PBMC H1 Primary T cell H2 Primary B cell H3 Primary Monocyte H4 HepG2 H5 3T3L1 H6 C2C12 H7 THP1 H8 Jurkat H9 Raji H10 ArgRS polypeptides were considered to be binding to a particular cell type if the mean cell bound fluorescence intensity was greater than 2 SD away from the reagent control values for that cell type.
- ArgRS1 N2 , ArgRS1 I1 , and ArgRS1 C2 appear to be major regulators of cytokine release, proliferation, and cell adhesion and chemotaxis.
- ArgRS1 N2 , ArgRS1 I1 , and ArgRS1 C2 appear to be major regulators of cytokine release, proliferation, and cell adhesion and chemotaxis.
- ArgRS1 C2 appear to be major regulators of cytokine release, proliferation, and cell adhesion and chemotaxis.
- the phenotypic screening data demonstrates that the ArgRS polypeptides ArgRS1 N2 (amino acids 1-71), ArgRS1 I1 (amino acids 168-530) and ArgRS1 C2 (amino acids 551-660) define the boundaries of a three novel protein domains that are highly active in a broad array of phenotypic screening assays.
- ArgRS polypeptides comprising amino acids 1-71, or 168-530, or 551-660 of Arginyl tRNA synthetase define the approximate boundaries (i.e. within about +/- 5 amino acids) of a three distinct novel, highly active ArgRS polypeptide domains, that are i) highly functionally active, ii) can be readily made and produced in E. coli, and iii) exhibit favorable protein stability and aggregation characteristics.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Hematology (AREA)
- Diabetes (AREA)
- General Engineering & Computer Science (AREA)
- Epidemiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Urology & Nephrology (AREA)
- Gastroenterology & Hepatology (AREA)
- Endocrinology (AREA)
- Neurology (AREA)
- Obesity (AREA)
- Biophysics (AREA)
- Cell Biology (AREA)
- Pathology (AREA)
- General Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
Claims (15)
- Eine therapeutische Zusammensetzung, umfassend ein isoliertes aminoacyl-tRNA-Synthetase (AARS)-Polypeptid, das mindestens 90%, 95%, 98% oder 100% identisch ist mit SEQ ID NO:29, 151 oder 137, wobei das Polypeptide eine extrazelluläre Signalaktivität und eine Löslichkeit von mindestens etwa 5 mg/ml aufweist, und wobei die Zusammensetzung eine Reinheit von mindestens etwa 95% auf Proteinbasis und weniger als etwa 10 EU Endotoxin/mg Protein aufweist.
- Ein isoliertes aminoacyl-tRNA-Synthetase (AARS)-Polypeptid, das mindestens 90%, 95%, 98% oder 100% identisch ist mit SEQ ID NO:29, 151 oder 137.
- Eine Zusammensetzung, umfassend ein isoliertes aminoacyl-tRNA-Synthetase (AARS)-Polypeptid von Anspruch 2, wobei das Polypeptid eine Löslichkeit von mindestens etwa 5 mg/ml aufweist, und wobei die Zusammensetzung eine Reinheit von mindestens etwa 95% auf Proteinbasis und weniger als etwa 10 EU Endotoxin/mg Protein aufweist.
- Die Zusammensetzung von Anspruch 3, wobei mindestens ein Rest oder ein festes Substrat kovalent oder nicht-kovalent mit jenem Polypeptid verbunden ist, oder wobei das AARS-Polypeptid mit einem heterologen Polypeptid fusioniert ist, wobei das heterologe Polypeptid mit dem N-Terminus des AARS-Polypeptids verbunden ist.
- Ein Antikörper, der eine Bindungsspezifität für ein isoliertes AARS-Polypeptid von Anspruch 2 aufweist, wobei die Affinität des Antikörpers für das AARS-Polypeptid etwa 10X oder mehr stärker ist als seine Affinität für ein entsprechendes Volllängen-AARS-Polypeptid.
- Eine Zusammensetzung, umfassend einen Antikörper von Anspruch 5, wobei der Antikörper eine Affinität von mindestens etwa 1 nM für das Polypeptid aufweist, und wobei der Antikörper eine nicht-kanonische Aktivität des AARS-Polypeptids antagonisiert.
- Eine Zusammensetzung, umfassend ein im Wesentlichen reines aminoacyl-tRNA-Synthetase (AARS)-Polypeptid von Anspruch 2 und ein Element ausgewählt aus der Gruppe bestehend aus(i) einem Bindungspartner, der das AARS-Polypeptid bindet,(ii) einer gentechnisch veränderter Zellpopulation, in der mindestens eine Zelle ein Polynukleotid umfasst, das für jenes AARS-Polypeptid kodiert, wobei die Zellen in der Lage sind, in einem serumfreien Medium zu wachsen,(iii) einer Zelle, die einen Zelloberflächenrezeptor oder einen extrazellulären Teil davon umfasst, der an das AARS-Polypeptid bindet, und ein Molekül von weniger als etwa 2000 Dalton oder ein zweites Polypeptid, das die Bindung oder die Wechselwirkung des AARS-Polypeptids und des extrazellulären Rezeptors moduliert,(iv) einer Zelle, die einen Zelloberflächenrezeptor oder einen extrazellulären Teil davon umfasst, der an das AARS-Polypeptid spezifisch bindet, wobei die Zelle ein Indikatormolekül umfasst, das die Detektion einer Veränderung in den Spiegeln oder Aktivität des Zelloberflächenrezeptors oder des extrazellulären Teils davon ermöglicht, und(v) einer gentechnisch veränderten Zellpopulation in der mindestens eine Zelle ein Polynukleotid umfasst, das jenes AARS-Polypeptid kodiert, mindestens etwa 10 Liter serumfreies Wachstumsmedium und einem sterilen Behälter.
- Ein isoliertes aminoacyl-tRNA-Synthetase (AARS)-Polypeptid, umfassend(i) eine Nukleotidsequenz, die ein AARS-Polypeptid von Anspruch 2 kodiert oder ein Komplement davon; oder(ii) eine Nukleotidsequenz, die spezifisch mit einer Nukleotidsequenz von Satz 1 hybridisiert.
- Ein Verfahren zur Bestimmung der Anwesenheit oder den Spiegeln eines AARS-Polypeptids in einer Probe, umfassend Kontaktieren der Probe mit ein oder mehr Bindungsmitteln, die spezifisch an ein AARS-Polypeptid von Anspruch 2 binden, Detektieren der Anwesenheit oder Abwesenheit des Bindungsmittels und dadurch Bestimmen der Anwesenheit oder der Spiegel des AARS-Polypeptids.
- Ein Verfahren zur Bestimmung der Anwesenheit oder den Spiegeln einer Polynukleotidsequenz einer AARS-Spleißvariante in einer Probe, umfassend Kontaktieren der Probe mit ein oder mehr Oligonukleotiden, die spezifisch mit einem Polynukleotid von Anspruch 8, Satz 1 hybridisieren, Detektieren der Anwesenheit oder Abwesenheit der Oligonukleotide in der Probe und dadurch Bestimmen der Anwesenheit oder der Spiegel der Polynukleotidsequenz der AARS-Spleißvariante.
- Ein Verfahren zur Identifizierung einer Verbindung , die spezifisch an ein aminoacyl-tRNA-Synthetase (AARS)-Polypeptid von Anspruch 2 bindet, umfassend,a) Kombinieren des AARS-Polypeptids mit mindestens einer Testverbindung unter geeigneten Bedingungen, undb) Detektieren der Bindung des AARS-Polypeptids an die Testverbindung, und dadurch Identifizieren einer Verbindung , die spezifisch an das AARS-Polypeptid bindet.
- Eine pharmazeutische Zusammensetzung, umfassend ein AARS-Polypeptid von Anspruch 2, ein Antikörper von Anspruch 5 oder ein AARS-Polynukleotid von Anspruch 8, und einen pharmazeutisch annehmbaren Träger.
- Ein in vitro Verfahren zur Modulation einer Zellaktivität einer Zelle oder eines Proteins, umfassend Kontaktieren der Zelle oder des Proteins mit einem AARS-Polypeptid von Anspruch 2, einem Antikörper von Anspruch 5 oder einem AARS-Polynukleotid von Anspruch 8 oder einer pharmazeutischen Zusammensetzung von Anspruch 12.
- Ein gentechnisch verändertes Volllängen-aminoacyl-tRNA Synthetase (AARS)-Protein umfassend eine heterologe Proteolysestelle, um die proteolytische Herstellung eines AARS-Polypeptids von Anspruch 2 zu ermöglichen.
- Eine therapeutische Zusammensetzung von Anspruch 1 oder ein AARS-Polypeptid von Anspruch 2 oder eine Zusammensetzung von Anspruch 3 oder 4 zur Verwendung in der Behandlung einer neurologischen Erkrankung oder für die humane mesenchymale Stammzell (MSC)-Transplantation, wobei wahlweise die neurologische Erkrankung ausgewählt ist von Neuroinflammation, Neurodegeneration und Hirnischämie.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US33079010P | 2010-05-03 | 2010-05-03 | |
US33077810P | 2010-05-03 | 2010-05-03 | |
US33078510P | 2010-05-03 | 2010-05-03 | |
PCT/US2011/034838 WO2011139986A2 (en) | 2010-05-03 | 2011-05-02 | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-trna synthetases |
Publications (3)
Publication Number | Publication Date |
---|---|
EP2566515A2 EP2566515A2 (de) | 2013-03-13 |
EP2566515A4 EP2566515A4 (de) | 2013-09-18 |
EP2566515B1 true EP2566515B1 (de) | 2017-08-02 |
Family
ID=44904419
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP11778118.7A Active EP2566515B1 (de) | 2010-05-03 | 2011-05-02 | Innovative entdeckung von therapeutischen und diagnostischen mitteln sowie antikörperzusammensetzungen im zusammenhang mit proteinfragmenten von arginyl-trna-synthetasen |
Country Status (7)
Country | Link |
---|---|
US (3) | US8961961B2 (de) |
EP (1) | EP2566515B1 (de) |
JP (2) | JP5976638B2 (de) |
CN (1) | CN103096925A (de) |
AU (1) | AU2011248355B2 (de) |
CA (1) | CA2797277C (de) |
WO (1) | WO2011139986A2 (de) |
Families Citing this family (33)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2403864B1 (de) | 2009-02-27 | 2015-08-12 | Atyr Pharma, Inc. | Polypeptidstrukturmotive im zusammenhang mit zellsignalisierungsaktivität |
JP6066900B2 (ja) | 2010-04-26 | 2017-01-25 | エータイアー ファーマ, インコーポレイテッド | システイニルtRNA合成酵素のタンパク質フラグメントに関連した治療用、診断用および抗体組成物の革新的発見 |
CN108165537A (zh) | 2010-04-27 | 2018-06-15 | Atyr医药公司 | 与苏氨酰-tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
AU2011248614B2 (en) | 2010-04-27 | 2017-02-16 | Pangu Biopharma Limited | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases |
EP2563911B1 (de) | 2010-04-28 | 2021-07-21 | aTyr Pharma, Inc. | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von alanyl-trna-synthetasen |
CN103118693B (zh) | 2010-04-29 | 2017-05-03 | Atyr 医药公司 | 与缬氨酰‑tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
EP2563912B1 (de) | 2010-04-29 | 2018-09-05 | aTyr Pharma, Inc. | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von asparaginyl-trna-synthetasen |
CA2797977C (en) | 2010-05-03 | 2019-08-20 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-trna synthetases |
WO2011139988A2 (en) | 2010-05-03 | 2011-11-10 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of seryl-trna synthetases |
US8981045B2 (en) | 2010-05-03 | 2015-03-17 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-tRNA synthetases |
EP2566515B1 (de) | 2010-05-03 | 2017-08-02 | aTyr Pharma, Inc. | Innovative entdeckung von therapeutischen und diagnostischen mitteln sowie antikörperzusammensetzungen im zusammenhang mit proteinfragmenten von arginyl-trna-synthetasen |
AU2011248101B2 (en) | 2010-05-04 | 2016-10-20 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex |
EP2566492B1 (de) | 2010-05-04 | 2016-10-26 | aTyr Pharma, Inc. | Innovative entdeckung therapeutischer zusammensetzungen im zusammenhang mit proteinfragmenten von glutamyl-prolyl-trna-synthetasen |
WO2011143482A2 (en) | 2010-05-14 | 2011-11-17 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-trna synthetases |
CN103096914B (zh) | 2010-05-17 | 2015-08-12 | Atyr医药公司 | 与亮氨酰-tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
JP6046607B2 (ja) | 2010-05-27 | 2016-12-21 | エータイアー ファーマ, インコーポレイテッド | グルタミニルtRNA合成酵素のタンパク質フラグメントに関連した治療用、診断用および抗体組成物の革新的発見 |
AU2011261486B2 (en) | 2010-06-01 | 2017-02-23 | Pangu Biopharma Limited | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of lysyl-tRNA synthetases |
KR20130102534A (ko) | 2010-07-12 | 2013-09-17 | 에이티와이알 파마, 인코포레이티드 | 히스티딜trna 합성효소의 단백질 단편에 관련된 치료적, 진단적, 및 항체 조성물의 혁신적 발견 |
WO2012021247A2 (en) | 2010-07-12 | 2012-02-16 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-trna synthetases |
KR102099730B1 (ko) | 2010-07-12 | 2020-04-16 | 에이티와이알 파마, 인코포레이티드 | 아스파르틸trna 합성효소의 단백질 단편에 관련된 치료적, 진단적, 및 항체 조성물의 혁신적 발견 |
US8999321B2 (en) | 2010-07-12 | 2015-04-07 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases |
CN103108650A (zh) | 2010-08-25 | 2013-05-15 | Atyr医药公司 | 与酪氨酰-tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
AU2011311956C1 (en) | 2010-10-06 | 2017-06-15 | Pangu Biopharma Limited | Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases |
WO2012158945A2 (en) * | 2011-05-19 | 2012-11-22 | The Scripps Research Institute | Compositions and methods for treating charcot-marie-tooth diseases and related neuronal diseases |
US9714419B2 (en) | 2011-08-09 | 2017-07-25 | Atyr Pharma, Inc. | PEGylated tyrosyl-tRNA synthetase polypeptides |
WO2013086216A1 (en) | 2011-12-06 | 2013-06-13 | Atyr Pharma, Inc. | Improved aspartyl-trna synthetases |
US9822353B2 (en) | 2011-12-06 | 2017-11-21 | Atyr Pharma, Inc. | PEGylated aspartyl-tRNA synthetase polypeptides |
AU2012368189B2 (en) | 2011-12-29 | 2017-08-31 | Atyr Pharma, Inc. | Aspartyl-tRNA synthetase-Fc conjugates |
EP2970921B1 (de) | 2013-03-15 | 2018-09-19 | aTyr Pharma, Inc. | Histidyl-trna-synthetase-fc-konjugate |
WO2015183667A1 (en) | 2014-05-28 | 2015-12-03 | The Regents Of The University Of California | HYBRID tRNA/pre-miRNA MOLECULES AND METHODS OF USE |
US10422003B2 (en) | 2015-03-23 | 2019-09-24 | The Regents Of The University Of California | Methods for detection of RNase activity |
AU2018256435A1 (en) | 2017-04-20 | 2019-11-07 | Atyr Pharma, Inc. | Compositions and methods for treating lung inflammation |
CN111343970A (zh) | 2017-09-13 | 2020-06-26 | 北卡罗莱纳州立大学 | 微针贴片局部诱导脂肪组织褐变治疗肥胖症 |
Family Cites Families (356)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US3773919A (en) | 1969-10-23 | 1973-11-20 | Du Pont | Polylactide-drug mixtures |
US4483105A (en) | 1983-03-11 | 1984-11-20 | Maschinenfabrik Gehring, Gesellschaft Mit Beschrankter Haftung & Co. Kommanditgesellschaft | Device to hold workpieces for the honing of their axial center bore |
US5506337A (en) | 1985-03-15 | 1996-04-09 | Antivirals Inc. | Morpholino-subunit combinatorial library and method |
CA1268404A (en) | 1985-03-15 | 1990-05-01 | Antivirals Inc. | Polynucleotide assay reagent and method |
US5217866A (en) | 1985-03-15 | 1993-06-08 | Anti-Gene Development Group | Polynucleotide assay reagent and method |
US5521063A (en) | 1985-03-15 | 1996-05-28 | Antivirals Inc. | Polynucleotide reagent containing chiral subunits and methods of use |
US5166315A (en) | 1989-12-20 | 1992-11-24 | Anti-Gene Development Group | Sequence-specific binding polymers for duplex nucleic acids |
US5034506A (en) | 1985-03-15 | 1991-07-23 | Anti-Gene Development Group | Uncharged morpholino-based polymers having achiral intersubunit linkages |
US4965188A (en) | 1986-08-22 | 1990-10-23 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme |
US4683202A (en) | 1985-03-28 | 1987-07-28 | Cetus Corporation | Process for amplifying nucleic acid sequences |
US4683195A (en) | 1986-01-30 | 1987-07-28 | Cetus Corporation | Process for amplifying, detecting, and/or-cloning nucleic acid sequences |
US4800159A (en) | 1986-02-07 | 1989-01-24 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences |
US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
DE3788914T2 (de) | 1986-09-08 | 1994-08-25 | Ajinomoto Kk | Verbindungen zur Spaltung von RNS an eine spezifische Position, Oligomere, verwendet bei der Herstellung dieser Verbindungen und Ausgangsprodukte für die Synthese dieser Oligomere. |
US4873192A (en) | 1987-02-17 | 1989-10-10 | The United States Of America As Represented By The Department Of Health And Human Services | Process for site specific mutagenesis without phenotypic selection |
US5091513A (en) | 1987-05-21 | 1992-02-25 | Creative Biomolecules, Inc. | Biosynthetic antibody binding sites |
US5132405A (en) | 1987-05-21 | 1992-07-21 | Creative Biomolecules, Inc. | Biosynthetic antibody binding sites |
US5766960A (en) | 1987-07-27 | 1998-06-16 | Australian Membrane And Biotechnology Research Institute | Receptor membranes |
ATE151467T1 (de) | 1987-11-30 | 1997-04-15 | Univ Iowa Res Found | Durch modifikationen an der 3'-terminalen phosphodiesterbindung stabilisierte dna moleküle, ihre verwendung als nukleinsäuresonden sowie als therapeutische mittel zur hemmung der expression spezifischer zielgene |
US5403711A (en) | 1987-11-30 | 1995-04-04 | University Of Iowa Research Foundation | Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved |
US5543508A (en) | 1987-12-15 | 1996-08-06 | Gene Shears Pty. Limited | Ribozymes |
US4904584A (en) | 1987-12-23 | 1990-02-27 | Genetics Institute, Inc. | Site-specific homogeneous modification of polypeptides |
US5130238A (en) | 1988-06-24 | 1992-07-14 | Cangene Corporation | Enhanced nucleic acid amplification process |
US5223409A (en) | 1988-09-02 | 1993-06-29 | Protein Engineering Corp. | Directed evolution of novel binding proteins |
GB8822228D0 (en) | 1988-09-21 | 1988-10-26 | Southern E M | Support-bound oligonucleotides |
US5110596A (en) | 1988-12-13 | 1992-05-05 | Alza Corporation | Delivery system comprising means for delivering agent to livestock |
US5034229A (en) | 1988-12-13 | 1991-07-23 | Alza Corporation | Dispenser for increasing feed conversion of hog |
US5530101A (en) | 1988-12-28 | 1996-06-25 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US5856092A (en) | 1989-02-13 | 1999-01-05 | Geneco Pty Ltd | Detection of a nucleic acid sequence or a change therein |
US5256775A (en) | 1989-06-05 | 1993-10-26 | Gilead Sciences, Inc. | Exonuclease-resistant oligonucleotides |
US6040138A (en) | 1995-09-15 | 2000-03-21 | Affymetrix, Inc. | Expression monitoring by hybridization to high density oligonucleotide arrays |
US6309822B1 (en) | 1989-06-07 | 2001-10-30 | Affymetrix, Inc. | Method for comparing copy number of nucleic acid sequences |
US5871928A (en) | 1989-06-07 | 1999-02-16 | Fodor; Stephen P. A. | Methods for nucleic acid analysis |
US5925525A (en) | 1989-06-07 | 1999-07-20 | Affymetrix, Inc. | Method of identifying nucleotide differences |
US5547839A (en) | 1989-06-07 | 1996-08-20 | Affymax Technologies N.V. | Sequencing of surface immobilized polymers utilizing microflourescence detection |
US5800992A (en) | 1989-06-07 | 1998-09-01 | Fodor; Stephen P.A. | Method of detecting nucleic acids |
US5143854A (en) | 1989-06-07 | 1992-09-01 | Affymax Technologies N.V. | Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof |
DE69028257T2 (de) | 1989-07-11 | 1997-01-09 | Gen Probe Inc | Verfahren zur Vervielfältigung von Nukleinsäuresequenzen |
CA2020958C (en) | 1989-07-11 | 2005-01-11 | Daniel L. Kacian | Nucleic acid sequence amplification methods |
US5725871A (en) | 1989-08-18 | 1998-03-10 | Danbiosyst Uk Limited | Drug delivery compositions comprising lysophosphoglycerolipid |
US5707644A (en) | 1989-11-04 | 1998-01-13 | Danbiosyst Uk Limited | Small particle compositions for intranasal drug delivery |
US5804604A (en) | 1989-12-21 | 1998-09-08 | Biogen, Inc. | Tat-derived transport polypeptides and fusion proteins |
US5652122A (en) | 1989-12-21 | 1997-07-29 | Frankel; Alan | Nucleic acids encoding and methods of making tat-derived transport polypeptides |
US6316003B1 (en) | 1989-12-21 | 2001-11-13 | Whitehead Institute For Biomedical Research | Tat-derived transport polypeptides |
US5623065A (en) | 1990-08-13 | 1997-04-22 | Isis Pharmaceuticals, Inc. | Gapped 2' modified oligonucleotides |
US5955589A (en) | 1991-12-24 | 1999-09-21 | Isis Pharmaceuticals Inc. | Gapped 2' modified oligonucleotides |
US5149797A (en) | 1990-02-15 | 1992-09-22 | The Worcester Foundation For Experimental Biology | Method of site-specific alteration of rna and production of encoded polypeptides |
US5220007A (en) | 1990-02-15 | 1993-06-15 | The Worcester Foundation For Experimental Biology | Method of site-specific alteration of RNA and production of encoded polypeptides |
US5466468A (en) | 1990-04-03 | 1995-11-14 | Ciba-Geigy Corporation | Parenterally administrable liposome formulation comprising synthetic lipids |
US5811387A (en) | 1990-05-15 | 1998-09-22 | Chiron Corporation | Peptoid mixtures |
US6582908B2 (en) | 1990-12-06 | 2003-06-24 | Affymetrix, Inc. | Oligonucleotides |
US5399363A (en) | 1991-01-25 | 1995-03-21 | Eastman Kodak Company | Surface modified anticancer nanoparticles |
US5455166A (en) | 1991-01-31 | 1995-10-03 | Becton, Dickinson And Company | Strand displacement amplification |
US5936731A (en) | 1991-02-22 | 1999-08-10 | Applied Spectral Imaging Ltd. | Method for simultaneous detection of multiple fluorophores for in situ hybridization and chromosome painting |
EP0519596B1 (de) | 1991-05-17 | 2005-02-23 | Merck & Co. Inc. | Verfahren zur Verminderung der Immunogenität der variablen Antikörperdomänen |
US5714331A (en) | 1991-05-24 | 1998-02-03 | Buchardt, Deceased; Ole | Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility |
US5719262A (en) | 1993-11-22 | 1998-02-17 | Buchardt, Deceased; Ole | Peptide nucleic acids having amino acid side chains |
US5539082A (en) | 1993-04-26 | 1996-07-23 | Nielsen; Peter E. | Peptide nucleic acids |
DE4137649C2 (de) | 1991-11-15 | 1997-11-20 | Gerhard Dingler | Bauelement |
US5270184A (en) | 1991-11-19 | 1993-12-14 | Becton, Dickinson And Company | Nucleic acid target generation |
US5412087A (en) | 1992-04-24 | 1995-05-02 | Affymax Technologies N.V. | Spatially-addressable immobilization of oligonucleotides and other biological polymers on surfaces |
US5756353A (en) | 1991-12-17 | 1998-05-26 | The Regents Of The University Of California | Expression of cloned genes in the lung by aerosol-and liposome-based delivery |
DE9115660U1 (de) | 1991-12-18 | 1992-07-30 | Aventis Research & Technologies GmbH & Co KG, 65929 Frankfurt | L-Phenylalanyl-tRNA-Synthetase mit erweiterter Substratselektivität aus Mikroorganismen |
US5700922A (en) | 1991-12-24 | 1997-12-23 | Isis Pharmaceuticals, Inc. | PNA-DNA-PNA chimeric macromolecules |
US5541061A (en) | 1992-04-29 | 1996-07-30 | Affymax Technologies N.V. | Methods for screening factorial chemical libraries |
ES2105262T3 (es) | 1992-05-18 | 1997-10-16 | Minnesota Mining & Mfg | Dispositivo de suministro de farmaco a traves de las mucosas. |
US5652355A (en) | 1992-07-23 | 1997-07-29 | Worcester Foundation For Experimental Biology | Hybrid oligonucleotide phosphorothioates |
DE69334258D1 (de) | 1992-08-21 | 2009-02-26 | Univ Bruxelles | Immunoglobuline ohne Leichtkette |
US6765087B1 (en) | 1992-08-21 | 2004-07-20 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US6005079A (en) | 1992-08-21 | 1999-12-21 | Vrije Universiteit Brussels | Immunoglobulins devoid of light chains |
US5293050A (en) | 1993-03-25 | 1994-03-08 | International Business Machines Corporation | Semiconductor quantum dot light emitting/detecting devices |
EP0705271B1 (de) | 1993-06-25 | 2002-11-13 | Affymetrix, Inc. (a Delaware Corporation) | Hybridisierung und sequenzierung von nukleinsäuren |
US5858659A (en) | 1995-11-29 | 1999-01-12 | Affymetrix, Inc. | Polymorphism detection |
US5543158A (en) | 1993-07-23 | 1996-08-06 | Massachusetts Institute Of Technology | Biodegradable injectable nanoparticles |
US6045996A (en) | 1993-10-26 | 2000-04-04 | Affymetrix, Inc. | Hybridization assays on oligonucleotide arrays |
US5420109A (en) | 1993-11-12 | 1995-05-30 | Houghten Pharmaceuticals, Inc. | Cytokine restraining agents |
US5446090A (en) | 1993-11-12 | 1995-08-29 | Shearwater Polymers, Inc. | Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules |
AU691550B2 (en) | 1993-12-09 | 1998-05-21 | Thomas Jefferson University | Compounds and methods for site-directed mutations in eukaryotic cells |
US6090555A (en) | 1997-12-11 | 2000-07-18 | Affymetrix, Inc. | Scanned image alignment systems and methods |
KR100230718B1 (ko) | 1994-03-16 | 1999-11-15 | 다니엘 엘. 캐시앙, 헨리 엘. 노르호프 | 등온 가닥 변위 핵산 증폭법 |
US5648211A (en) | 1994-04-18 | 1997-07-15 | Becton, Dickinson And Company | Strand displacement amplification using thermophilic enzymes |
US5571639A (en) | 1994-05-24 | 1996-11-05 | Affymax Technologies N.V. | Computer-aided engineering system for design of sequence arrays and lithographic masks |
US5759833A (en) | 1994-05-27 | 1998-06-02 | Cubist Pharmaceuticals, Inc. | Human isoleucyl-tRNA synthetase proteins, nucleic acids and tester strains comprising same |
US6287850B1 (en) | 1995-06-07 | 2001-09-11 | Affymetrix, Inc. | Bioarray chip reaction apparatus and its manufacture |
US6379897B1 (en) | 2000-11-09 | 2002-04-30 | Nanogen, Inc. | Methods for gene expression monitoring on electronic microarrays |
NL9401150A (nl) | 1994-07-12 | 1996-02-01 | Nederland Ptt | Werkwijze voor het aan een ontvangzijde aanbieden van een van een zendzijde afkomstig eerste aantal videosignalen, alsmede systeem, alsmede zender, alsmede netwerk, en alsmede ontvanger. |
US5798240A (en) | 1994-09-13 | 1998-08-25 | Cubist Pharmaceuticals, Inc. | Recombinant mycobacterial methionyl-tRNA synthetase genes and methods of use therefore |
US5756327A (en) | 1994-09-13 | 1998-05-26 | Cubist Pharmaceuticals, Inc. | Recombinant mycobacterial isoleucyl-tRNA synthetase genes, tester strains and assays |
US5824784A (en) | 1994-10-12 | 1998-10-20 | Amgen Inc. | N-terminally chemically modified protein compositions and methods |
US5795716A (en) | 1994-10-21 | 1998-08-18 | Chee; Mark S. | Computer-aided visualization and analysis system for sequence evaluation |
WO1996014834A1 (en) | 1994-11-10 | 1996-05-23 | University Of Kentucky Research Foundation | Implantable refillable controlled release device to deliver drugs directly to an internal portion of the body |
US5545729A (en) | 1994-12-22 | 1996-08-13 | Hybridon, Inc. | Stabilized ribozyme analogs |
US5932462A (en) | 1995-01-10 | 1999-08-03 | Shearwater Polymers, Inc. | Multiarmed, monofunctional, polymer for coupling to molecules and surfaces |
US6169073B1 (en) | 1995-02-16 | 2001-01-02 | Bayer Corporation | Peptides and peptidomimetics with structural similarity to human p53 that activate p53 function |
IE80468B1 (en) | 1995-04-04 | 1998-07-29 | Elan Corp Plc | Controlled release biodegradable nanoparticles containing insulin |
US5801013A (en) | 1995-05-26 | 1998-09-01 | Cubist Pharmaceuticals, Inc. | Helicobacter aminoacyl-tRNA synthetase proteins, nucleic acids and strains comprising same |
US5545531A (en) | 1995-06-07 | 1996-08-13 | Affymax Technologies N.V. | Methods for making a device for concurrently processing multiple biological chip assays |
US5652356A (en) | 1995-08-17 | 1997-07-29 | Hybridon, Inc. | Inverted chimeric and hybrid oligonucleotides |
US5733729A (en) | 1995-09-14 | 1998-03-31 | Affymetrix, Inc. | Computer-aided probability base calling for arrays of nucleic acid probes on chips |
US5843655A (en) | 1995-09-18 | 1998-12-01 | Affymetrix, Inc. | Methods for testing oligonucleotide arrays |
US6300063B1 (en) | 1995-11-29 | 2001-10-09 | Affymetrix, Inc. | Polymorphism detection |
JP2001501447A (ja) * | 1996-01-11 | 2001-02-06 | コリクサ コーポレイション | 乳癌の処置および診断のための組成物および方法 |
EP1288298A3 (de) | 1996-01-19 | 2003-03-12 | Smithkline Beecham Plc | TRNA Synthetasen aus Staphylococcus aureus |
US5795757A (en) | 1997-01-17 | 1998-08-18 | Smithkline Beecham, P.L.C. | DNA encoding threonyl tRNA synthetase from staphylococcus aureus |
JPH11504820A (ja) | 1996-01-19 | 1999-05-11 | スミスクライン・ビーチャム・パブリック・リミテッド・カンパニー | スタフィロコッカス・アウレウスのスレオニル−tRNAシンセターゼ |
GB9601067D0 (en) | 1996-01-19 | 1996-03-20 | Smithkline Beecham Plc | Novel compounds |
EP0785267A1 (de) | 1996-01-19 | 1997-07-23 | Smithkline Beecham Plc | Valyl-TRNA Synthetase aus Staphylococcus Aureus |
EP0880598A4 (de) | 1996-01-23 | 2005-02-23 | Affymetrix Inc | Verfahren zur analyse von nukleinsäure |
US5837196A (en) | 1996-01-26 | 1998-11-17 | The Regents Of The University Of California | High density array fabrication and readout method for a fiber optic biosensor |
PL189137B1 (pl) | 1996-02-02 | 2005-06-30 | Alza Corp | Implantowane urządzenie do dostarczania substancji aktywnej |
US6395292B2 (en) | 1996-02-02 | 2002-05-28 | Alza Corporation | Sustained delivery of an active agent using an implantable system |
US6156331A (en) | 1996-02-02 | 2000-12-05 | Alza Corporation | Sustained delivery of an active agent using an implantable system |
AU2189397A (en) | 1996-02-08 | 1997-08-28 | Affymetrix, Inc. | Chip-based speciation and phenotypic characterization of microorganisms |
US6114122A (en) | 1996-03-26 | 2000-09-05 | Affymetrix, Inc. | Fluidics station with a mounting system and method of using |
US6238866B1 (en) | 1996-04-16 | 2001-05-29 | The United States Of America As Represented By The Secretary Of The Army | Detector for nucleic acid typing and methods of using the same |
GB9607991D0 (en) | 1996-04-18 | 1996-06-19 | Smithkline Beecham Plc | Novel compounds |
US5795758A (en) | 1997-04-18 | 1998-08-18 | Smithkline Beecham Corporation | DNA encoding histidyl tRNA synthetase variant from Streptococcus pneumoniae |
GB9607993D0 (en) | 1996-04-18 | 1996-06-19 | Smithkline Beecham Plc | Novel compounds |
US5976109A (en) | 1996-04-30 | 1999-11-02 | Medtronic, Inc. | Apparatus for drug infusion implanted within a living body |
GB9609262D0 (en) | 1996-05-02 | 1996-07-03 | Isis Innovation | Peptide library and method |
US5958342A (en) | 1996-05-17 | 1999-09-28 | Incyte Pharmaceuticals, Inc. | Jet droplet device |
KR100203919B1 (ko) | 1996-10-04 | 1999-06-15 | 신동권 | 수용성 단백질을 생산하는 새로운 발현 플라스미드 |
US5853993A (en) | 1996-10-21 | 1998-12-29 | Hewlett-Packard Company | Signal enhancement method and kit |
US5885815A (en) | 1996-11-01 | 1999-03-23 | Cubist Pharmaceuticals, Inc. | Candida isoleucyl-tRNA synthetase proteins, nucleic acids and strains comprising same |
WO1998020020A2 (en) | 1996-11-06 | 1998-05-14 | Sequenom, Inc. | High density immobilization of nucleic acids |
US5804386A (en) | 1997-01-15 | 1998-09-08 | Incyte Pharmaceuticals, Inc. | Sets of labeled energy transfer fluorescent primers and their use in multi component analysis |
US6309824B1 (en) | 1997-01-16 | 2001-10-30 | Hyseq, Inc. | Methods for analyzing a target nucleic acid using immobilized heterogeneous mixtures of oligonucleotide probes |
US5776749A (en) | 1997-01-17 | 1998-07-07 | Smithkline Beecham P.L.C. | DNA encoding cysteinyl tRNA synthetase from Staphylococcus aureus |
CA2289116A1 (en) | 1997-05-06 | 1998-11-12 | Human Genome Sciences, Inc. | Enterococcus faecalis polynucleotides and polypeptides |
WO1998056915A2 (en) | 1997-06-12 | 1998-12-17 | Research Corporation Technologies, Inc. | Artificial antibody polypeptides |
ATE319856T1 (de) | 1997-06-13 | 2006-03-15 | Affymetrix Inc A Delaware Corp | Verfahren zur erkennung von genpolymorphismen und allelexpression unter verwendung von sondenchips |
US6333179B1 (en) | 1997-06-20 | 2001-12-25 | Affymetrix, Inc. | Methods and compositions for multiplex amplification of nucleic acids |
US5882892A (en) | 1997-07-23 | 1999-03-16 | Smithkline Beecham Corporation | Asps |
US5858720A (en) | 1997-07-23 | 1999-01-12 | Smithkline Beecham Corporation | Hiss |
US5939298A (en) | 1997-07-23 | 1999-08-17 | Smithkline Beecham Corporation | DNA encoding phenylalanyl tRNA synthetase alpha sub-unit from chlamydi a trachomatis |
WO1999005574A1 (en) | 1997-07-25 | 1999-02-04 | Affymetrix, Inc. | Method and system for providing a probe array chip design database |
US6420108B2 (en) | 1998-02-09 | 2002-07-16 | Affymetrix, Inc. | Computer-aided display for comparative gene expression |
CA2299625A1 (en) | 1997-08-15 | 1999-02-25 | Affymetrix, Inc. | Polymorphism detection utilizing clustering analysis |
US6794499B2 (en) | 1997-09-12 | 2004-09-21 | Exiqon A/S | Oligonucleotide analogues |
US7572582B2 (en) | 1997-09-12 | 2009-08-11 | Exiqon A/S | Oligonucleotide analogues |
DE19742706B4 (de) | 1997-09-26 | 2013-07-25 | Pieris Proteolab Ag | Lipocalinmuteine |
US6511803B1 (en) | 1997-10-10 | 2003-01-28 | President And Fellows Of Harvard College | Replica amplification of nucleic acid arrays |
ATE291097T1 (de) | 1997-10-31 | 2005-04-15 | Affymetrix Inc A Delaware Corp | Expressionsprofile in adulten und fötalen organen |
US6054274A (en) | 1997-11-12 | 2000-04-25 | Hewlett-Packard Company | Method of amplifying the signal of target nucleic acid sequence analyte |
US6013449A (en) | 1997-11-26 | 2000-01-11 | The United States Of America As Represented By The Department Of Health And Human Services | Probe-based analysis of heterozygous mutations using two-color labelling |
PT1041975E (pt) | 1997-12-22 | 2003-01-31 | Alza Corp | Membranas de controlo de taxa para dispositivos de fornecimento controlado de drogas |
JP4494629B2 (ja) | 1997-12-30 | 2010-06-30 | インターシア セラピューティクス,インコーポレイティド | メンブレンプラグを備えた有益な作用剤の供給システム |
EP1051624A4 (de) | 1998-01-29 | 2002-05-02 | Glaucus Proteomics B V | Hochdichte matrizen zur proteomanalyse und verfahren und zusammensetzungen dafür |
EP1051523A2 (de) | 1998-02-02 | 2000-11-15 | Amersham Pharmacia Biotech AB | Nuklein-säure analyse verfahren |
US6083726A (en) | 1998-02-03 | 2000-07-04 | Lucent Technologies, Inc. | Methods for polynucleotide synthesis and articles for polynucleotide hybridization |
AU2583899A (en) | 1998-02-04 | 1999-08-23 | Invitrogen Corporation | Microarrays and uses therefor |
US6428960B1 (en) | 1998-03-04 | 2002-08-06 | Onyx Pharmaceuticals, Inc. | Selection method for producing recombinant baculovirus |
JP2002505110A (ja) | 1998-03-04 | 2002-02-19 | オニックス ファーマシューティカルズ,インコーポレイティド | 遺伝物質の高生産性発現のためのバキュロウイルス発現系及び方法 |
AUPP221098A0 (en) | 1998-03-06 | 1998-04-02 | Diatech Pty Ltd | V-like domain binding molecules |
US6020135A (en) | 1998-03-27 | 2000-02-01 | Affymetrix, Inc. | P53-regulated genes |
US6004755A (en) | 1998-04-07 | 1999-12-21 | Incyte Pharmaceuticals, Inc. | Quantitative microarray hybridizaton assays |
US6284497B1 (en) | 1998-04-09 | 2001-09-04 | Trustees Of Boston University | Nucleic acid arrays and methods of synthesis |
WO2002083898A1 (en) | 2001-04-18 | 2002-10-24 | Genset S.A. | Full-length human cdnas encoding potentially secreted proteins |
US6525185B1 (en) | 1998-05-07 | 2003-02-25 | Affymetrix, Inc. | Polymorphisms associated with hypertension |
US6268210B1 (en) | 1998-05-27 | 2001-07-31 | Hyseq, Inc. | Sandwich arrays of biological compounds |
WO1999067284A2 (en) | 1998-06-20 | 1999-12-29 | Washington University | Membrane-permeant peptide complexes for medical imaging, diagnostics, and pharmaceutical therapy |
US7306784B2 (en) | 1998-06-20 | 2007-12-11 | Washington University | Membrane-permeant peptide complexes for medical imaging, diagnostics, and pharmaceutical therapy |
US6589503B1 (en) | 1998-06-20 | 2003-07-08 | Washington University | Membrane-permeant peptide complexes for medical imaging, diagnostics, and pharmaceutical therapy |
US6255090B1 (en) | 1998-07-15 | 2001-07-03 | E. I. Du Pont De Nemours & Company | Plant aminoacyl-tRNA synthetase |
US6271441B1 (en) | 1998-07-21 | 2001-08-07 | E. I. Du Pont De Nemours & Company | Plant aminoacyl-tRNA synthetase |
US6306643B1 (en) | 1998-08-24 | 2001-10-23 | Affymetrix, Inc. | Methods of using an array of pooled probes in genetic analysis |
US6185561B1 (en) | 1998-09-17 | 2001-02-06 | Affymetrix, Inc. | Method and apparatus for providing and expression data mining database |
US6203989B1 (en) | 1998-09-30 | 2001-03-20 | Affymetrix, Inc. | Methods and compositions for amplifying detectable signals in specific binding assays |
US6316193B1 (en) | 1998-10-06 | 2001-11-13 | Origene Technologies, Inc. | Rapid-screen cDNA library panels |
US6262216B1 (en) | 1998-10-13 | 2001-07-17 | Affymetrix, Inc. | Functionalized silicon compounds and methods for their synthesis and use |
EP1124990B1 (de) | 1998-10-27 | 2006-01-18 | Affymetrix, Inc. | Komplexitätsmanagement und analyse genomischer dna |
US6696619B1 (en) | 1998-11-05 | 2004-02-24 | Omolayo O. Famodu | Plant aminoacyl-tRNA synthetases |
US6263287B1 (en) | 1998-11-12 | 2001-07-17 | Scios Inc. | Systems for the analysis of gene expression data |
US6309828B1 (en) | 1998-11-18 | 2001-10-30 | Agilent Technologies, Inc. | Method and apparatus for fabricating replicate arrays of nucleic acid molecules |
US6210922B1 (en) | 1998-11-30 | 2001-04-03 | National Research Council Of Canada | Serum free production of recombinant proteins and adenoviral vectors |
US7115396B2 (en) | 1998-12-10 | 2006-10-03 | Compound Therapeutics, Inc. | Protein scaffolds for antibody mimics and other binding proteins |
US6245518B1 (en) | 1998-12-11 | 2001-06-12 | Hyseq, Inc. | Polynucleotide arrays and methods of making and using the same |
US6351712B1 (en) | 1998-12-28 | 2002-02-26 | Rosetta Inpharmatics, Inc. | Statistical combining of cell expression profiles |
US6586802B2 (en) | 1999-01-14 | 2003-07-01 | Mitsubishi Denki Kabushiki Kaisha | Semiconductor device |
US6312906B1 (en) | 1999-01-15 | 2001-11-06 | Imperial College Innovations, Ltd. | Immobilized nucleic acid hybridization reagent and method |
US6251601B1 (en) | 1999-02-02 | 2001-06-26 | Vysis, Inc. | Simultaneous measurement of gene expression and genomic abnormalities using nucleic acid microarrays |
US6329145B1 (en) | 1999-02-09 | 2001-12-11 | Gilead Science, Inc. | Determining non-nucleic acid molecule binding to target by competition with nucleic acid ligand |
US6177248B1 (en) | 1999-02-24 | 2001-01-23 | Affymetrix, Inc. | Downstream genes of tumor suppressor WT1 |
AU773291B2 (en) | 1999-03-10 | 2004-05-20 | Government of The United States of America, as represented by The Secretary Department of Health & Human Services, The National Institutes of Health, The | UPA, a universal protein array system |
US7084125B2 (en) | 1999-03-18 | 2006-08-01 | Exiqon A/S | Xylo-LNA analogues |
EP1183387A2 (de) | 1999-04-09 | 2002-03-06 | Arcturus Engineering, Inc. | Generischer cdna oder proteinmatrix für angepasste tests |
US6284465B1 (en) | 1999-04-15 | 2001-09-04 | Agilent Technologies, Inc. | Apparatus, systems and method for locating nucleic acids bound to surfaces |
IL146126A0 (en) | 1999-04-30 | 2002-07-25 | Slil Biomedical Corp | Novel polyamine analog conjugates and quinone conjugates as therapies for cancers and prostate diseases |
ES2283298T3 (es) | 1999-05-04 | 2007-11-01 | Santaris Pharma A/S | Analogos de l-ribo-lna. |
US6268141B1 (en) | 1999-05-12 | 2001-07-31 | Beckman Coulter, Inc. | Immobilization of unmodified biopolymers to acyl fluoride activated substrates |
US6605115B1 (en) | 1999-06-05 | 2003-08-12 | Board Of Trustees Of The Leland Stanford Junior University | Method and composition for inhibiting cardiovascular cell proliferation |
WO2001007628A2 (en) * | 1999-07-22 | 2001-02-01 | Incyte Genomics, Inc. | Human synthetases |
US6387620B1 (en) | 1999-07-28 | 2002-05-14 | Gilead Sciences, Inc. | Transcription-free selex |
AU6770800A (en) | 1999-08-13 | 2001-03-13 | Yale University | Analysis of sequence tags with hairpin primers |
US6669951B2 (en) | 1999-08-24 | 2003-12-30 | Cellgate, Inc. | Compositions and methods for enhancing drug delivery across and into epithelial tissues |
US7229961B2 (en) | 1999-08-24 | 2007-06-12 | Cellgate, Inc. | Compositions and methods for enhancing drug delivery across and into ocular tissues |
AU7473500A (en) | 1999-09-01 | 2001-03-26 | University Of Pittsburgh | Identification of peptides that facilitate uptake and cytoplasmic and/or nucleartransport of proteins, dna and viruses |
WO2001019999A1 (fr) | 1999-09-14 | 2001-03-22 | Shanghai Biorigin Gene Development Co. Ltd. | Gene codant une nouvelle threonyl-arnt synthase, ses utilisations et procedes de preparation |
US6171797B1 (en) | 1999-10-20 | 2001-01-09 | Agilent Technologies Inc. | Methods of making polymeric arrays |
US6548060B1 (en) | 1999-11-18 | 2003-04-15 | Sunghoon Kim | Anti-apoptotic use of human glutaminyl-tRNA synthetase with two consecutive pro-apoptotic mediators |
US6372431B1 (en) | 1999-11-19 | 2002-04-16 | Incyte Genomics, Inc. | Mammalian toxicological response markers |
DE19957827C2 (de) | 1999-11-25 | 2003-06-12 | Epigenomics Ag | Verwendung eines Oligomer-Arrays mit PNA- und/oder DNA-Oligomeren auf einer Oberfläche |
US6383749B2 (en) | 1999-12-02 | 2002-05-07 | Clontech Laboratories, Inc. | Methods of labeling nucleic acids for use in array based hybridization assays |
US6283949B1 (en) | 1999-12-27 | 2001-09-04 | Advanced Cardiovascular Systems, Inc. | Refillable implantable drug delivery pump |
IL151047A0 (en) | 2000-02-03 | 2003-04-10 | Res Dev Foundation | Signaling aptamers that transduce molecular recognition to a differential signal |
US6267152B1 (en) | 2000-02-18 | 2001-07-31 | Wisconsin Label Corporation | Hang tag and method of applying hang tag to an elongated object |
JP3464460B2 (ja) | 2000-02-24 | 2003-11-10 | ジー・ケー・エヌ・レブロ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング | ドライブシャフト |
CN1311300A (zh) | 2000-03-02 | 2001-09-05 | 上海博德基因开发有限公司 | 一种新的多肽-人苏氨酰-tRNA合成酶14和编码这种多肽的多核苷酸 |
US6376191B1 (en) | 2000-03-22 | 2002-04-23 | Mergen, Ltd. | Microarray-based analysis of polynucleotide sequence variations |
ATE473275T1 (de) | 2000-03-31 | 2010-07-15 | Scripps Research Inst | Menschliche aminoacyl-trna synthetase polypeptide zur regulation der angiogenese |
US7273844B2 (en) | 2000-03-31 | 2007-09-25 | The Scripps Research Institute | Tryptophanyl-tRNA synthetase-derived polypeptides useful for the regulation of angiogenesis |
US6436703B1 (en) | 2000-03-31 | 2002-08-20 | Hyseq, Inc. | Nucleic acids and polypeptides |
US7144984B2 (en) | 2000-03-31 | 2006-12-05 | The Scripps Research Institute | Human aminoacyl-tRNA synthetase polypeptides useful for the regulation of angiogenesis |
WO2001075178A2 (en) | 2000-04-04 | 2001-10-11 | Enanta Pharmaceuticals, Inc. | Methods for identifying peptide aptamers capable of altering a cell phenotype |
CN1322818A (zh) | 2000-05-09 | 2001-11-21 | 上海博德基因开发有限公司 | 一种新的多肽——人II类氨酰基-tRNA合成酶10和编码这种多肽的多核苷酸 |
US20040181830A1 (en) | 2001-05-07 | 2004-09-16 | Kovalic David K. | Nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement |
WO2001088162A2 (en) | 2000-05-16 | 2001-11-22 | Genway Biotech, Inc. | Methods and vectors for generating antibodies in avian species and uses therefor |
JP2004512014A (ja) | 2000-05-18 | 2004-04-22 | 学校法人日本大学 | 虚血状態の検査方法 |
WO2001090330A2 (en) | 2000-05-25 | 2001-11-29 | Incyte Genomics, Inc. | AMINOACYL tRNA SYNTHETASES |
US20020055125A1 (en) | 2000-06-05 | 2002-05-09 | Chiron Corporation | Microarrays for performing proteomic analyses |
US6531283B1 (en) | 2000-06-20 | 2003-03-11 | Molecular Staging, Inc. | Protein expression profiling |
US6386749B1 (en) | 2000-06-26 | 2002-05-14 | Affymetrix, Inc. | Systems and methods for heating and mixing fluids |
KR100787073B1 (ko) | 2000-06-28 | 2007-12-21 | 글리코파이, 인크. | 변형된 당단백질의 제조방법 |
US6380377B1 (en) | 2000-07-14 | 2002-04-30 | Applied Gene Technologies, Inc. | Nucleic acid hairpin probes and uses thereof |
JP2002071687A (ja) | 2000-08-31 | 2002-03-12 | Canon Inc | 変異遺伝子のスクリーニング方法 |
WO2002031463A2 (en) | 2000-08-31 | 2002-04-18 | Motorola, Inc. | High density column and row addressable electrode arrays |
CN1341725A (zh) | 2000-09-07 | 2002-03-27 | 上海博德基因开发有限公司 | 一种新的多肽——人苏氨酰-tRNA合成酶48.73和编码这种多肽的多核苷酸 |
CN1341727A (zh) | 2000-09-07 | 2002-03-27 | 上海博德基因开发有限公司 | 一种新的多肽——甲硫氨酰tRNA合成酶35.09和编码这种多肽的多核苷酸 |
AU2002218166A1 (en) | 2000-09-08 | 2002-03-22 | Universitat Zurich | Collections of repeat proteins comprising repeat modules |
AU2001292959A1 (en) | 2000-09-22 | 2002-04-02 | Clontech Laboratories, Inc. | Highly sensitive proteomic analysis methods and kits and systems for practicing the same |
US6376190B1 (en) | 2000-09-22 | 2002-04-23 | Somalogic, Inc. | Modified SELEX processes without purified protein |
US6596541B2 (en) | 2000-10-31 | 2003-07-22 | Regeneron Pharmaceuticals, Inc. | Methods of modifying eukaryotic cells |
CN1352242A (zh) | 2000-11-02 | 2002-06-05 | 上海博德基因开发有限公司 | 一种新的多肽——人谷氨酰tRNA合成酶12.65和编码这种多肽的多核苷酸 |
CN1352252A (zh) | 2000-11-06 | 2002-06-05 | 上海博德基因开发有限公司 | 一种新的多肽——人II类氨酰基-tRNA合成酶11.77和编码这种多肽的多核苷酸 |
EP1337504A4 (de) | 2000-11-08 | 2005-10-05 | Cellgate Inc | Neue polyaminanalogon-aminosäure-konjugate zur verwendung als krebsmittel |
US6436386B1 (en) | 2000-11-14 | 2002-08-20 | Shearwater Corporation | Hydroxyapatite-targeting poly (ethylene glycol) and related polymers |
AU2002223349A1 (en) | 2000-11-28 | 2002-06-11 | Mcgill University | Incorporation and priming function of trnalys in hiv and related viruses |
JP2004537270A (ja) | 2000-12-15 | 2004-12-16 | インサイト・ゲノミックス・インコーポレイテッド | アミノアシルtRNA合成酵素 |
US20020183936A1 (en) | 2001-01-24 | 2002-12-05 | Affymetrix, Inc. | Method, system, and computer software for providing a genomic web portal |
MXPA03007358A (es) | 2001-02-16 | 2004-12-13 | Cellgate Inc | Transportadores que contienen porciones de arginina separadas. |
KR100869914B1 (ko) | 2001-02-23 | 2008-11-21 | 더 스크립스 리서치 인스티튜트 | 혈관 형성 조절에 유용한 트립토파닐-tRNA 신세타제기원 폴리펩타이드 |
US6903189B2 (en) | 2001-03-21 | 2005-06-07 | The Scripps Research Institute | Human aminoacyl-tRNA synthetase polypeptides useful for the regulation of angiogenesis |
WO2002085923A2 (en) | 2001-04-19 | 2002-10-31 | The Scripps Research Institute | In vivo incorporation of unnatural amino acids |
ATE472609T1 (de) | 2001-04-26 | 2010-07-15 | Amgen Mountain View Inc | Kombinatorische bibliotheken von monomerdomänen |
US20050089932A1 (en) | 2001-04-26 | 2005-04-28 | Avidia Research Institute | Novel proteins with targeted binding |
US20050048512A1 (en) | 2001-04-26 | 2005-03-03 | Avidia Research Institute | Combinatorial libraries of monomer domains |
US20050053973A1 (en) | 2001-04-26 | 2005-03-10 | Avidia Research Institute | Novel proteins with targeted binding |
US20040175756A1 (en) | 2001-04-26 | 2004-09-09 | Avidia Research Institute | Methods for using combinatorial libraries of monomer domains |
US20030215827A1 (en) | 2001-05-22 | 2003-11-20 | Henry Yue | Aminoacyl trna synthetases |
KR100405919B1 (ko) | 2001-06-05 | 2003-11-14 | 주식회사 이매진 | p43의 N-말단 펩타이드를 유효성분으로 하는 면역증강용 약학조성물 |
US20040018505A1 (en) | 2001-06-29 | 2004-01-29 | Lee Ernestine A. | Aminoacyl trna synthetases |
US6632611B2 (en) | 2001-07-20 | 2003-10-14 | Affymetrix, Inc. | Method of target enrichment and amplification |
US6872529B2 (en) | 2001-07-25 | 2005-03-29 | Affymetrix, Inc. | Complexity management of genomic DNA |
AU2002332430A1 (en) | 2001-07-26 | 2003-02-17 | Novartis Ag | Methods of treating neuropilin-mediated diseases |
JP2005514005A (ja) | 2001-09-04 | 2005-05-19 | エクシコン エ/エス | 新規のlna組成物およびその使用 |
US7785827B2 (en) | 2001-09-20 | 2010-08-31 | University Of Houston System | Method and composition for leucyl-tRNA synthetases and derivatives thereof that activate and aminoacylate non-leucine amino acid to tRNA adaptor molecules |
WO2003050072A1 (en) | 2001-12-07 | 2003-06-19 | Slil Biomedical Corporation | Cycloalkyl substituted polyamines for cancer therapy and methods of synthesis therefor |
EP1461084A2 (de) | 2001-12-11 | 2004-09-29 | The Board Of Trustees Of The Leland Stanford Junior University | Guanidinium transport-reagenzien und konjugate |
US20040048290A1 (en) | 2001-12-13 | 2004-03-11 | Lee Ernestine A | Aminoacyl trna synthetases |
US7011945B2 (en) | 2001-12-21 | 2006-03-14 | Eastman Kodak Company | Random array of micro-spheres for the analysis of nucleic acids |
KR100464261B1 (ko) | 2002-01-24 | 2005-01-03 | 주식회사 파나진 | Pna 올리고머를 합성하기 위한 신규한 단량체 및 그의제조방법 |
US7244592B2 (en) | 2002-03-07 | 2007-07-17 | Dyax Corp. | Ligand screening and discovery |
KR20030084444A (ko) | 2002-04-26 | 2003-11-01 | 주식회사 파나진 | Pna 올리고머를 합성하기 위한 신규한 단량체 및 그의제조방법 |
US7569575B2 (en) | 2002-05-08 | 2009-08-04 | Santaris Pharma A/S | Synthesis of locked nucleic acid derivatives |
WO2003094862A2 (en) | 2002-05-13 | 2003-11-20 | Rigel Pharmaceuticals, Inc. | tRNA SYNTHASE: MODULATORS OF ANGIOGENESIS |
SE0201863D0 (en) | 2002-06-18 | 2002-06-18 | Cepep Ab | Cell penetrating peptides |
JPWO2004022736A1 (ja) | 2002-08-30 | 2005-12-22 | 独立行政法人科学技術振興機構 | 遺伝子のターゲティング破壊法および超耐熱菌ゲノム、ならびにこれらを利用したゲノムチップ |
DK1534830T3 (da) * | 2002-09-06 | 2011-02-14 | Isogenica Ltd | In vito-peptid-ekspressionsbibliotek |
US7011949B2 (en) | 2002-09-30 | 2006-03-14 | Agilent Technologies, Inc. | Methods and compositions for producing labeled probe nucleic acids for use in array based comparative genomic hybridization applications |
CA2500687A1 (en) | 2002-10-02 | 2004-04-15 | Genentech, Inc. | Compositions and methods for the diagnosis and treatment of tumor |
US7028629B2 (en) | 2003-02-03 | 2006-04-18 | Richard Walcome | Self-sealing port light assembly |
KR100575251B1 (ko) | 2003-03-03 | 2006-05-02 | 재단법인서울대학교산학협력재단 | p38/JTV-1을 유효성분으로 하는 암 치료용 약학적조성물 및 암 치료용 약학적 조성물의 스크리닝 방법 |
WO2004087875A2 (en) | 2003-03-25 | 2004-10-14 | Incyte Corporation | Nucleic acid-associated proteins |
CA2523651A1 (en) | 2003-05-01 | 2005-02-03 | Replidyne, Inc. | Antibacterial methods and compositions |
EP3604537B1 (de) | 2003-06-13 | 2021-12-08 | Alnylam Europe AG | Doppelsträngige ribonukleinsäure mit erhöhter wirksamkeit in einem organismus |
DE602004027770D1 (de) | 2003-07-07 | 2010-07-29 | Scripps Research Inst | Zusammensetzungen aus orthogonalen Lysyl-TRNA- und Aminoacyl-TRNA-Synthetasepaaren und deren Verwendung |
US20060160175A1 (en) | 2003-07-07 | 2006-07-20 | The Scripps Research Institute | Compositions of orthogonal leucyl-trna and aminoacyl-trna synthetase pairs and uses thereof |
US7211668B2 (en) | 2003-07-28 | 2007-05-01 | Panagene, Inc. | PNA monomer and precursor |
US7431915B2 (en) | 2003-10-31 | 2008-10-07 | The Regents Of The University Of California | Peptides whose uptake by cells is controllable |
US20080220049A1 (en) | 2003-12-05 | 2008-09-11 | Adnexus, A Bristol-Myers Squibb R&D Company | Compositions and methods for intraocular delivery of fibronectin scaffold domain proteins |
US7741071B2 (en) | 2003-12-18 | 2010-06-22 | The Scripps Research Institute | Selective incorporation of 5-hydroxytryptophan into proteins in mammalian cells |
KR100599454B1 (ko) | 2004-04-27 | 2006-07-12 | 재단법인서울대학교산학협력재단 | 종양 억제자로 작용하는 aim3의 신규 용도 |
CA2562685C (en) | 2004-04-27 | 2013-09-17 | Alnylam Pharmaceuticals, Inc. | Single-stranded and double-stranded oligonucleotides comprising a 2-arylpropyl moiety |
PL2484365T3 (pl) | 2004-06-04 | 2014-03-31 | Scripps Research Inst | Kompozycje i sposoby leczenia chorób neowaskularnych |
WO2006088490A2 (en) | 2004-06-30 | 2006-08-24 | Alnylam Pharmaceuticals, Inc. | Oligonucleotides comprising a non-phosphate backbone linkage |
AU2005328382C1 (en) | 2004-07-21 | 2013-01-24 | Alnylam Pharmaceuticals, Inc. | Oligonucleotides comprising a modified or non-natural nucleobase |
US20060079673A1 (en) | 2004-08-02 | 2006-04-13 | Paul Glidden | Polynucleotides encoding tRNA synthetase fragments and uses thereof |
US20060024288A1 (en) | 2004-08-02 | 2006-02-02 | Pfizer Inc. | tRNA synthetase fragments |
US8282921B2 (en) | 2004-08-02 | 2012-10-09 | Paul Glidden | tRNA synthetase fragments |
US20060078553A1 (en) | 2004-10-07 | 2006-04-13 | Paul Glidden | Diverse multi-unit complexes including a tRNA synthetase fragment |
EP1913011B1 (de) | 2004-08-04 | 2016-11-02 | Alnylam Pharmaceuticals Inc. | Oligonukleotide mit einem an einer modifizierten oder nicht-natürlichen nukleobase angebundenen liganden |
US7459529B2 (en) | 2004-11-24 | 2008-12-02 | Seoul National University Industry Foundation | AIMP2-DX2 and its uses |
US8003780B2 (en) | 2004-11-24 | 2011-08-23 | Neomics Co., Ltd. | AIMP2-DX2 gene and SiRNA targeting AIMP2-DX2 |
US7939496B2 (en) | 2004-12-22 | 2011-05-10 | Ambrx, Inc. | Modified human growth horomone polypeptides and their uses |
AU2005319518B2 (en) | 2004-12-22 | 2010-09-09 | Ambrx, Inc. | Compositions of aminoacyl-tRNA synthetase and uses thereof |
CN101273140B (zh) | 2005-08-18 | 2013-01-16 | Ambrx公司 | tRNA组合物和其用途 |
US7514229B2 (en) | 2005-09-29 | 2009-04-07 | The Board Of Trustees Of The Leland Stanford Junior University | Methods for diagnosing and evaluating treatment of blood disorders |
AU2006320361B2 (en) | 2005-12-02 | 2012-10-25 | The Scripps Research Institute | Angiogenic tyrosyl tRNA synthetase compositions and methods |
PT1999259E (pt) | 2006-03-03 | 2014-09-24 | California Inst Of Techn | Incorporação sítio-específica de aminoácidos em moléculas |
US20070232677A1 (en) | 2006-03-14 | 2007-10-04 | Marton Laurence J | Treatment and prevention of vascular hyperplasia using polyamine and polyamine analog compounds |
WO2007111993A2 (en) | 2006-03-22 | 2007-10-04 | Cellgate, Inc. | Polyamine analogs as therapeutic agents for skin diseases |
AU2007268364B8 (en) | 2006-05-26 | 2013-05-16 | Obodies Limited | OB fold domains |
EP2052088A2 (de) | 2006-08-02 | 2009-04-29 | Genizon Biosciences | Genkarte der mit psoriasis assoziierten menschlichen gene |
EP2520935A3 (de) | 2006-08-09 | 2013-02-13 | Homestead Clinical Corporation | Organspezifische Proteine und Verfahren zu deren Verwendung |
EP2064316B1 (de) | 2006-09-08 | 2012-01-25 | Ambrx, Inc. | Standortspezifische inkorporation nicht natürlicher aminosäuren in wirbeltierzellen |
AU2007321817A1 (en) | 2006-11-17 | 2008-05-22 | Novartis Ag | Lingo binding molecules and pharmaceutical use thereof |
WO2008063113A1 (en) | 2006-11-20 | 2008-05-29 | Cepep Iii Ab | Cell -penetrating peptides and constructs containing them consisting 15-25 amino acids of tumor supressor protein p14arf or p19arf |
US20100138941A1 (en) | 2007-04-27 | 2010-06-03 | Imagene Co., Ltd. | Method for screening immune modulator |
JP2009017840A (ja) | 2007-07-13 | 2009-01-29 | Japan Agengy For Marine-Earth Science & Technology | 外来遺伝子を細胞に安定に保持する方法 |
WO2009059056A2 (en) | 2007-11-02 | 2009-05-07 | The Scripps Research Institute | A genetically encoded boronate amino acid |
JP5585904B2 (ja) | 2008-02-08 | 2014-09-10 | 独立行政法人理化学研究所 | アミノアシルtRNA合成酵素活性を有するポリペプチド及びその利用 |
US20110104139A1 (en) | 2008-03-11 | 2011-05-05 | The University Of North Carolina At Chapel Hill | ANGIOSTATIC COMPOSITIONS COMPRISING TRUNCATED TYROSYL-tRNA SYNTHETASE POLYPEPTIDES AND METHODS OF USING SAME |
JP5771142B2 (ja) | 2008-06-11 | 2015-08-26 | エータイアー ファーマ, インコーポレイテッド | チロシル−tRNAシンテターゼポリペプチドの血小板新生活性 |
EP2310498A1 (de) | 2008-06-26 | 2011-04-20 | Atyr Pharma, Inc. | Glycyl-trna-synthetasen mit nichtkanonischen biologischen aktivitäten umfassende zusammensetzungen und verfahren |
CA2734892A1 (en) | 2008-08-18 | 2010-02-25 | Seoul National University Industry Foundation | Method for controlling cancer metastasis or cancer cell migration by modulating the cellular level of lysyl trna synthetase |
KR101067817B1 (ko) | 2008-10-10 | 2011-09-27 | 서울대학교산학협력단 | Aimp1 폴리펩티드에 대한 항체를 포함하는 관절염 진단용 조성물 |
EP2351575B1 (de) | 2008-10-10 | 2016-12-14 | SNU R & DB Foundation | Grs in der verwendung zur behandlung von krebs |
KR101067815B1 (ko) | 2009-02-05 | 2011-09-27 | 서울대학교산학협력단 | 제1형 당뇨병의 신규한 진단 마커 |
WO2010096170A2 (en) | 2009-02-19 | 2010-08-26 | President And Fellows Of Harvard College | Inhibition of trna synthetases and therapeutic applications thereof |
EP2403864B1 (de) | 2009-02-27 | 2015-08-12 | Atyr Pharma, Inc. | Polypeptidstrukturmotive im zusammenhang mit zellsignalisierungsaktivität |
WO2010107825A2 (en) | 2009-03-16 | 2010-09-23 | Pangu Biopharma Limited | Compositions and methods comprising histidyl-trna synthetase splice variants having non-canonical biological activities |
EP2414513B1 (de) | 2009-03-31 | 2015-10-28 | Atyr Pharma, Inc. | Zusammensetzungen und verfahren mit aspartyl-trna-synthetasen mit nichtkanonischen biologischen aktivitäten |
US20110150885A1 (en) | 2009-12-11 | 2011-06-23 | Atyr Pharma, Inc. | Aminoacyl trna synthetases for modulating hematopoiesis |
AU2010327926B2 (en) | 2009-12-11 | 2014-05-08 | Atyr Pharma, Inc. | Aminoacyl tRNA synthetases for modulating inflammation |
US8828395B2 (en) | 2009-12-11 | 2014-09-09 | Atyr Pharma, Inc. | Antibodies that bind tyrosyl-tRNA synthetases |
US8012804B1 (en) | 2009-12-23 | 2011-09-06 | Western Digital (Fremont), Llc | Method and system for mounting lasers on energy assisted magnetic recording heads |
US8828685B2 (en) | 2010-02-04 | 2014-09-09 | The Scripps Research Institute | Monomeric forms of human aminoacyl-t-RNA synthetases having non-canonical biological activities |
JP6066900B2 (ja) | 2010-04-26 | 2017-01-25 | エータイアー ファーマ, インコーポレイテッド | システイニルtRNA合成酵素のタンパク質フラグメントに関連した治療用、診断用および抗体組成物の革新的発見 |
AU2011248614B2 (en) | 2010-04-27 | 2017-02-16 | Pangu Biopharma Limited | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases |
CN108165537A (zh) | 2010-04-27 | 2018-06-15 | Atyr医药公司 | 与苏氨酰-tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
EP2563911B1 (de) | 2010-04-28 | 2021-07-21 | aTyr Pharma, Inc. | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von alanyl-trna-synthetasen |
EP2563912B1 (de) | 2010-04-29 | 2018-09-05 | aTyr Pharma, Inc. | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von asparaginyl-trna-synthetasen |
CN103118693B (zh) | 2010-04-29 | 2017-05-03 | Atyr 医药公司 | 与缬氨酰‑tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
EP2566515B1 (de) | 2010-05-03 | 2017-08-02 | aTyr Pharma, Inc. | Innovative entdeckung von therapeutischen und diagnostischen mitteln sowie antikörperzusammensetzungen im zusammenhang mit proteinfragmenten von arginyl-trna-synthetasen |
CA2797977C (en) | 2010-05-03 | 2019-08-20 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-trna synthetases |
US8981045B2 (en) | 2010-05-03 | 2015-03-17 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-tRNA synthetases |
WO2011139988A2 (en) | 2010-05-03 | 2011-11-10 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of seryl-trna synthetases |
AU2011248101B2 (en) | 2010-05-04 | 2016-10-20 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex |
EP2566492B1 (de) | 2010-05-04 | 2016-10-26 | aTyr Pharma, Inc. | Innovative entdeckung therapeutischer zusammensetzungen im zusammenhang mit proteinfragmenten von glutamyl-prolyl-trna-synthetasen |
WO2011143482A2 (en) | 2010-05-14 | 2011-11-17 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-trna synthetases |
CN103096914B (zh) | 2010-05-17 | 2015-08-12 | Atyr医药公司 | 与亮氨酰-tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
JP6046607B2 (ja) | 2010-05-27 | 2016-12-21 | エータイアー ファーマ, インコーポレイテッド | グルタミニルtRNA合成酵素のタンパク質フラグメントに関連した治療用、診断用および抗体組成物の革新的発見 |
AU2011261486B2 (en) | 2010-06-01 | 2017-02-23 | Pangu Biopharma Limited | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of lysyl-tRNA synthetases |
KR102099730B1 (ko) | 2010-07-12 | 2020-04-16 | 에이티와이알 파마, 인코포레이티드 | 아스파르틸trna 합성효소의 단백질 단편에 관련된 치료적, 진단적, 및 항체 조성물의 혁신적 발견 |
WO2012021247A2 (en) | 2010-07-12 | 2012-02-16 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-trna synthetases |
US8999321B2 (en) | 2010-07-12 | 2015-04-07 | Atyr Pharma, Inc. | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases |
KR20130102534A (ko) | 2010-07-12 | 2013-09-17 | 에이티와이알 파마, 인코포레이티드 | 히스티딜trna 합성효소의 단백질 단편에 관련된 치료적, 진단적, 및 항체 조성물의 혁신적 발견 |
CN103108650A (zh) | 2010-08-25 | 2013-05-15 | Atyr医药公司 | 与酪氨酰-tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现 |
AU2011311956C1 (en) | 2010-10-06 | 2017-06-15 | Pangu Biopharma Limited | Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases |
WO2012158945A2 (en) | 2011-05-19 | 2012-11-22 | The Scripps Research Institute | Compositions and methods for treating charcot-marie-tooth diseases and related neuronal diseases |
US9714419B2 (en) | 2011-08-09 | 2017-07-25 | Atyr Pharma, Inc. | PEGylated tyrosyl-tRNA synthetase polypeptides |
US9822353B2 (en) | 2011-12-06 | 2017-11-21 | Atyr Pharma, Inc. | PEGylated aspartyl-tRNA synthetase polypeptides |
WO2013086216A1 (en) | 2011-12-06 | 2013-06-13 | Atyr Pharma, Inc. | Improved aspartyl-trna synthetases |
AU2012368189B2 (en) | 2011-12-29 | 2017-08-31 | Atyr Pharma, Inc. | Aspartyl-tRNA synthetase-Fc conjugates |
DK2814514T3 (en) | 2012-02-16 | 2017-12-18 | Atyr Pharma Inc | Histidyl tRNA synthetases for the treatment of autoimmune and inflammatory diseases |
US20140066321A1 (en) | 2012-07-23 | 2014-03-06 | Pangu Biopharma Limited | Structures of human histidyl-trna synthetase and methods of use |
-
2011
- 2011-05-02 EP EP11778118.7A patent/EP2566515B1/de active Active
- 2011-05-02 CA CA2797277A patent/CA2797277C/en active Active
- 2011-05-02 WO PCT/US2011/034838 patent/WO2011139986A2/en active Application Filing
- 2011-05-02 US US13/696,042 patent/US8961961B2/en active Active
- 2011-05-02 AU AU2011248355A patent/AU2011248355B2/en active Active
- 2011-05-02 CN CN2011800308106A patent/CN103096925A/zh active Pending
- 2011-05-02 JP JP2013509149A patent/JP5976638B2/ja active Active
-
2015
- 2015-01-09 US US14/593,620 patent/US9593322B2/en active Active
- 2015-09-16 JP JP2015183033A patent/JP2015231389A/ja not_active Withdrawn
-
2016
- 2016-10-24 US US15/332,495 patent/US20170165362A1/en not_active Abandoned
Non-Patent Citations (1)
Title |
---|
None * |
Also Published As
Publication number | Publication date |
---|---|
WO2011139986A3 (en) | 2012-03-01 |
US9593322B2 (en) | 2017-03-14 |
CA2797277C (en) | 2021-02-23 |
JP5976638B2 (ja) | 2016-08-23 |
CA2797277A1 (en) | 2011-11-10 |
US20170165362A1 (en) | 2017-06-15 |
EP2566515A2 (de) | 2013-03-13 |
AU2011248355A1 (en) | 2012-12-13 |
JP2013529075A (ja) | 2013-07-18 |
CN103096925A (zh) | 2013-05-08 |
US20150252348A1 (en) | 2015-09-10 |
JP2015231389A (ja) | 2015-12-24 |
AU2011248355B2 (en) | 2017-01-19 |
EP2566515A4 (de) | 2013-09-18 |
US8961961B2 (en) | 2015-02-24 |
WO2011139986A2 (en) | 2011-11-10 |
US20130142774A1 (en) | 2013-06-06 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10563192B2 (en) | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases | |
US10220080B2 (en) | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases | |
US20200140571A1 (en) | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-trna synthetases | |
US10563191B2 (en) | Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases | |
EP2566515B1 (de) | Innovative entdeckung von therapeutischen und diagnostischen mitteln sowie antikörperzusammensetzungen im zusammenhang mit proteinfragmenten von arginyl-trna-synthetasen | |
EP2563381B1 (de) | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von isoleucyl-trna-synthetasen | |
EP2593126B1 (de) | Innovative erkennung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen in zusammenhang mit proteinfragmenten von histidyl-trna-synthetasen | |
US20190062454A1 (en) | Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-trna synthetase | |
EP2566516B1 (de) | Innovative entdeckung von therapeutischen und diagnostischen mitteln sowie antikörperzusammensetzungen im zusammenhang mit proteinfragmenten von seryl-trna-synthetasen | |
EP2566499B1 (de) | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten aus einem p38-multi-trna-synthetase-komplex | |
EP2593124B1 (de) | Innovative erkennung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen in zusammenhang mit proteinfragmenten von aspartyl-trna-synthetasen | |
EP2566495B1 (de) | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von phenylalanyl-alpha-trna-synthetasen | |
EP2575856B1 (de) | Therapeutische, diagnostische und antikörperhaltige zusammensetzungen in zusammenhang mit proteinfragmenten von glutaminyl-trna-synthetasen | |
EP2563911B1 (de) | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von alanyl-trna-synthetasen | |
EP2563383B1 (de) | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von valyl-trna-synthetasen | |
EP2608801B1 (de) | Innovative erkennung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen in zusammenhang mit proteinfragmenten von tyrosyl-trna-synthetasen | |
EP2563912B1 (de) | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von asparaginyl-trna-synthetasen | |
EP2593125B1 (de) | Innovative erkennung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen in zusammenhang mit proteinfragmenten von glycyl-trna-synthetasen | |
EP2566492B1 (de) | Innovative entdeckung therapeutischer zusammensetzungen im zusammenhang mit proteinfragmenten von glutamyl-prolyl-trna-synthetasen | |
US20160010075A1 (en) | INNOVATIVE DISCOVERY OF THERAPEUTIC, DIAGNOSTIC, AND ANTIBODY COMPOSITIONS RELATED TO PROTEIN FRAGMENTS OF LEUCYL-tRNA SYNTHETASES | |
EP2571513B1 (de) | Innovative entdeckung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen im zusammenhang mit proteinfragmenten von leucyl-trna-synthetasen |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
17P | Request for examination filed |
Effective date: 20121121 |
|
AK | Designated contracting states |
Kind code of ref document: A2 Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR |
|
AX | Request for extension of the european patent |
Extension state: BA ME |
|
RIN1 | Information on inventor provided before grant (corrected) |
Inventor name: GREENE, LESLIE ANN Inventor name: VASSEROT, ALAIN P. Inventor name: WATKINS, JEFFRY D. Inventor name: QUINN, CHERYL L. Inventor name: CHIANG, KYLE P. Inventor name: MENDLEIN, JOHN D. Inventor name: LO, WING-SZE Inventor name: HONG, FEI |
|
A4 | Supplementary search report drawn up and despatched |
Effective date: 20130819 |
|
RIC1 | Information provided on ipc code assigned before grant |
Ipc: A61K 39/395 20060101AFI20130812BHEP Ipc: A61P 37/00 20060101ALI20130812BHEP Ipc: A61K 39/39 20060101ALI20130812BHEP Ipc: A61K 38/43 20060101ALI20130812BHEP Ipc: A61K 38/16 20060101ALI20130812BHEP Ipc: C12N 9/00 20060101ALI20130812BHEP |
|
17Q | First examination report despatched |
Effective date: 20140724 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: EXAMINATION IS IN PROGRESS |
|
GRAP | Despatch of communication of intention to grant a patent |
Free format text: ORIGINAL CODE: EPIDOSNIGR1 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: GRANT OF PATENT IS INTENDED |
|
INTG | Intention to grant announced |
Effective date: 20170222 |
|
GRAS | Grant fee paid |
Free format text: ORIGINAL CODE: EPIDOSNIGR3 |
|
GRAA | (expected) grant |
Free format text: ORIGINAL CODE: 0009210 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE PATENT HAS BEEN GRANTED |
|
AK | Designated contracting states |
Kind code of ref document: B1 Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR |
|
AX | Request for extension of the european patent |
Extension state: BA ME |
|
REG | Reference to a national code |
Ref country code: GB Ref legal event code: FG4D |
|
REG | Reference to a national code |
Ref country code: CH Ref legal event code: EP Ref country code: AT Ref legal event code: REF Ref document number: 913748 Country of ref document: AT Kind code of ref document: T Effective date: 20170815 |
|
REG | Reference to a national code |
Ref country code: IE Ref legal event code: FG4D |
|
REG | Reference to a national code |
Ref country code: DE Ref legal event code: R096 Ref document number: 602011040195 Country of ref document: DE |
|
REG | Reference to a national code |
Ref country code: NL Ref legal event code: MP Effective date: 20170802 |
|
REG | Reference to a national code |
Ref country code: AT Ref legal event code: MK05 Ref document number: 913748 Country of ref document: AT Kind code of ref document: T Effective date: 20170802 |
|
REG | Reference to a national code |
Ref country code: LT Ref legal event code: MG4D |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: LT Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: AT Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: FI Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: SE Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: HR Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: NL Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: NO Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20171102 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: ES Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: PL Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: GR Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20171103 Ref country code: RS Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: IS Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20171202 Ref country code: LV Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: BG Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20171102 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: DK Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: CZ Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: RO Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
REG | Reference to a national code |
Ref country code: DE Ref legal event code: R097 Ref document number: 602011040195 Country of ref document: DE |
|
REG | Reference to a national code |
Ref country code: FR Ref legal event code: PLFP Year of fee payment: 8 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: EE Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: SK Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: SM Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 Ref country code: IT Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
PLBE | No opposition filed within time limit |
Free format text: ORIGINAL CODE: 0009261 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT |
|
26N | No opposition filed |
Effective date: 20180503 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: SI Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
REG | Reference to a national code |
Ref country code: CH Ref legal event code: PL |
|
REG | Reference to a national code |
Ref country code: BE Ref legal event code: MM Effective date: 20180531 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: MC Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
REG | Reference to a national code |
Ref country code: IE Ref legal event code: MM4A |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: CH Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES Effective date: 20180531 Ref country code: LI Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES Effective date: 20180531 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: LU Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES Effective date: 20180502 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: IE Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES Effective date: 20180502 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: BE Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES Effective date: 20180531 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: MT Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES Effective date: 20180502 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: TR Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: HU Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO Effective date: 20110502 Ref country code: PT Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: MK Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES Effective date: 20170802 Ref country code: CY Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
PG25 | Lapsed in a contracting state [announced via postgrant information from national office to epo] |
Ref country code: AL Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT Effective date: 20170802 |
|
P01 | Opt-out of the competence of the unified patent court (upc) registered |
Effective date: 20230426 |
|
PGFP | Annual fee paid to national office [announced via postgrant information from national office to epo] |
Ref country code: GB Payment date: 20240527 Year of fee payment: 14 |
|
PGFP | Annual fee paid to national office [announced via postgrant information from national office to epo] |
Ref country code: DE Payment date: 20240530 Year of fee payment: 14 |
|
PGFP | Annual fee paid to national office [announced via postgrant information from national office to epo] |
Ref country code: FR Payment date: 20240527 Year of fee payment: 14 |